header logo image


Page 5«..4567..1020..»

Archive for the ‘Stem Cell Negative’ Category

Data Evaluating Tafasitamab with and without Lenalidomide in Combination with R-CHOP in Patients with DLBCL Presented at ASH 2020 – Business Wire

Saturday, December 12th, 2020

WILMINGTON, Del. & PLANEGG/MUNICH, Germany--(BUSINESS WIRE)--Incyte (Nasdaq:INCY) and MorphoSys AG (FSE: MOR; Prime Standard Segment; MDAX & TecDAX; NASDAQ:MOR) announce that preliminary data from firstMIND, the ongoing Phase 1b, open-label, randomized study on the safety and efficacy of tafasitamab or tafasitamab plus lenalidomide in addition to R-CHOP for patients with newly diagnosed diffuse large B-cell lymphoma (DLBCL) were presented today during the 62nd American Society of Hematology Annual Meeting & Exposition (ASH). Additionally, a long-term subgroup analysis of the L-MIND study investigating tafasitamab combined with lenalidomide in patients with relapsed or refractory DLBCL was also presented at ASH.

The preliminary results of firstMIND indicate that tafasitamab plus lenalidomide in addition to R-CHOP shows an acceptable tolerability profile. Toxicities appear to be similar to what is expected with R-CHOP alone or in combination with lenalidomide. Serious or severe neutropenia and thrombocytopenia events (grade 3 or higher) were more frequent in the tafasitamab plus lenalidomide arm. The incidence of febrile neutropenia was comparable between both arms and the average relative dose intensity of R-CHOP was maintained in both arms. Interim response assessments after three cycles were available for 45 patients. In both arms combined, 41/45 (91.1%) of patients had an objective response as per Lugano 20141.

The preliminary data from this ongoing study in first-line DLBCL warrant further investigation. To that end, MorphoSys and Incyte plan to initiate frontMIND, a Phase 3 trial evaluating tafasitamab plus lenalidomide in combination with R-CHOP compared to R-CHOP alone as first-line treatment for patients with newly diagnosed DLBCL.

The initial results of the firstMIND study, shared today at ASH, as well as the long-term analyses from L-MIND, underscore the potential of tafasitamab as a combination therapeutic for patients with DLBCL, where there remains a significant unmet need. Along with our partners at MorphoSys, we are pleased to be moving forward with the initiation of a Phase 3 study in 2021, said Steven Stein, M.D., Chief Medical Officer at Incyte.

The preliminary firstMIND study results mark another important step as we explore the potential of tafasitamab as a backbone therapy, said Dr. Malte Peters, Chief Research and Development Officer at MorphoSys. Given the data available to date, including data from the L-MIND study, we believe that the mechanism of action, efficacy and safety profile of tafasitamab have the potential to make it a preferred combination partner as we seek to transform the standard of care in DLBCL. We are committed to developing innovative therapies to battle this aggressive disease for the benefit of patients with DLBCL, and look forward to beginning the planned frontMIND in the first half of 2021.

In addition to the firstMIND data presented today, the long-term L-MIND analyses showed that treatment with tafasitamab plus lenalidomide resulted in durable responses after 2 years of follow-up. At the time of analysis, patients with complete responses (CR) continued to experience durable treatment responses, including long duration of response (DoR) and overall survival (OS). The data also showed that tafasitamab plus lenalidomide taken for 12 cycles, followed by tafasitamab until progression, did not result in any unexpected safety signals2.

In July 2020, the FDA approved Monjuvi (tafasitamab-cxix), a humanized Fc-modified cytolytic CD19-targeting monoclonal antibody, in combination with lenalidomide for the treatment of adult patients with relapsed or refractory DLBCL not otherwise specified, including DLBCL arising from low grade lymphoma, and who are not eligible for autologous stem cell transplant (ASCT). This indication is approved under accelerated approval based on overall response rate. Continued approval for this indication may be contingent upon verification and description of clinical benefit in a confirmatory trial(s)3.

The FDA decision represented the first approval of a second-line treatment for adult patients with DLBCL who progressed during or after first-line therapy.

About Diffuse Large B-cell Lymphoma (DLBCL)

DLBCL is the most common type of non-Hodgkin lymphoma in adults worldwide4, characterized by rapidly growing masses of malignant B-cells in the lymph nodes, spleen, liver, bone marrow or other organs. It is an aggressive disease with about one in three patients not responding to initial therapy or relapsing thereafter5. In the United States each year, approximately 10,000 patients are diagnosed with relapsed or refractory DLBCL who are not eligible for autologous stem cell transplant (ASCT)6,7,8.

About firstMIND

The firstMIND (NCT04134936) trial is a Phase 1b, randomized study of tafasitamab + R-CHOP (Arm A) or tafasitamab + lenalidomide + R-CHOP (Arm B) in patients with newly diagnosed diffuse large B-cell lymphoma (DLBCL). The study includes a safety run-in phase and a main phase. In the safety run-in phase, 24 patients were enrolled. The primary objective is to assess safety; secondary objectives include objective response rate, PET negative complete response (PET-CR) rate at end of treatment, progression-free survival, event-free survival, long-term safety, pharmacokinetics and immunogenicity of tafasitamab.

About Tafasitamab

Tafasitamab is a humanized Fc-modified cytolytic CD19 targeting monoclonal antibody. In 2010, MorphoSys licensed exclusive worldwide rights to develop and commercialize tafasitamab from Xencor, Inc. Tafasitamab incorporates an XmAb engineered Fc domain, which mediates B-cell lysis through apoptosis and immune effector mechanism including antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP).

Monjuvi (tafasitamab-cxix) is approved by the U.S. Food and Drug Administration in combination with lenalidomide for the treatment of adult patients with relapsed or refractory diffuse large B-cell lymphoma (DLBCL) not otherwise specified, including DLBCL arising from low grade lymphoma, and who are not eligible for autologous stem cell transplant (ASCT). This indication is approved under accelerated approval based on overall response rate. Continued approval for this indication may be contingent upon verification and description of clinical benefit in a confirmatory trial(s).

In January 2020, MorphoSys and Incyte entered into a collaboration and licensing agreement to further develop and commercialize tafasitamab globally. Monjuvi is being co-commercialized by Incyte and MorphoSys in the United States. Incyte has exclusive commercialization rights outside the United States.

A marketing authorization application (MAA) seeking the approval of tafasitamab in combination with lenalidomide in the EU has been validated by the European Medicines Agency (EMA) and is currently under review for the treatment of adult patients with relapsed or refractory DLBCL, including DLBCL arising from low grade lymphoma, who are not candidates for ASCT.

Tafasitamab is being clinically investigated as a therapeutic option in B-cell malignancies in a number of ongoing combination trials.

Monjuvi is a registered trademark of MorphoSys AG.

XmAb is a registered trademark of Xencor, Inc.

Important Safety Information

What are the possible side effects of MONJUVI?

MONJUVI may cause serious side effects, including:

The most common side effects of MONJUVI include:

These are not all the possible side effects of MONJUVI.

Call your doctor for medical advice about side effects. You may report side effects to FDA at 1-800-FDA-1088.

Before you receive MONJUVI, tell your healthcare provider about all your medical conditions, including if you:

You should also read the lenalidomide Medication Guide for important information about pregnancy, contraception, and blood and sperm donation.

Tell your healthcare provider about all the medications you take, including prescription and over-the-counter medicines, vitamins, and herbal supplements.

Please see the full Prescribing Information for Monjuvi, including Patient Information, for additional Important Safety Information.

About Incyte

Incyte is a Wilmington, Delaware-based, global biopharmaceutical company focused on finding solutions for serious unmet medical needs through the discovery, development and commercialization of proprietary therapeutics. For additional information on Incyte, please visit Incyte.com and follow @Incyte.

About MorphoSys

MorphoSys (FSE & NASDAQ: MOR) is a commercial-stage biopharmaceutical company dedicated to the discovery, development and commercialization of exceptional, innovative therapies for patients suffering from serious diseases. The focus is on cancer. Based on its leading expertise in antibody, protein and peptide technologies, MorphoSys, together with its partners, has developed and contributed to the development of more than 100 product candidates, of which 27 are currently in clinical development. In 2017, Tremfya, developed by Janssen Research & Development, LLC and marketed by Janssen Biotech, Inc., for the treatment of plaque psoriasis, became the first drug based on MorphoSys antibody technology to receive regulatory approval. In July 2020, the U.S. Food and Drug Administration (FDA) granted accelerated approval of MorphoSys proprietary product Monjuvi (tafasitamab-cxix) in combination with lenalidomide in patients with a certain type of lymphoma.

Headquartered near Munich, Germany, the MorphoSys group, including the fully owned U.S. subsidiary MorphoSys US Inc., has ~500 employees. More information at http://www.morphosys.com or http://www.morphosys-us.com.

Monjuvi is a registered trademark of MorphoSys AG.

Tremfya is a registered trademark of Janssen Biotech, Inc.

Incyte Forward-Looking Statements

Except for the historical information set forth herein, the matters set forth in this press release - including statements about: plans to initiate frontMIND, a Phase 3 trial evaluating tafasitamab plus lenalidomide in combination with R-CHOP compared to R-CHOP alone as first-line treatment for patients with newly diagnosed DLBC; whether the mechanism of action, efficacy and safety profile of tafasitamab have the potential to make it a preferred or ideal combination partner in the treatment of DLBCL and, whether it will change or become the standard of care for the treatment of DLBCL; whether and when, if ever, confirmatory trials of tafasitamab will result in the conditional FDA approval of tafasitamab in the conditionally approved indication described above becoming a final approval; whether and when, if ever, the EMA will approve the filed MAA for tafasitamab; and additional development of tafasitamab, including in B-cell malignancies - contain predictions, estimates and other forward-looking statements.

These forward-looking statements are based on the Incytes current expectations and subject to risks and uncertainties that may cause actual results to differ materially, including unanticipated developments in and risks related to: unanticipated delays; further research and development and the results of clinical trials possibly being unsuccessful or insufficient to meet applicable regulatory standards or warrant continued development; the ability to enroll sufficient numbers of subjects in clinical trials; determinations made by the FDA or the EMA; clinical and commercial supply of products in development or being commercialized; Incytes dependence on its relationships with its collaboration partners; the efficacy or safety of Incytes products and the products of its collaboration partners; the acceptance of Incytes products and the products of its collaboration partners in the marketplace; market competition; sales, marketing, manufacturing and distribution requirements; greater than expected expenses; expenses relating to litigation or strategic activities; and other risks detailed from time to time in Incytes reports filed with the Securities and Exchange Commission, including its quarterly report on Form 10-Q for the quarter ended September 30, 2020. Incyte disclaims any intent or obligation to update these forward-looking statements.

MorphoSys Forward-Looking Statements

This communication contains certain forward-looking statements concerning the MorphoSys group of companies, including the expectations regarding Monjuvis ability to treat patients with relapsed or refractory diffuse large B-cell lymphoma, the further clinical development of tafasitamab-cxix, including ongoing confirmatory trials, additional interactions with regulatory authorities and expectations regarding future regulatory filings and possible additional approvals for tafasitamab-cxix as well as the commercial performance of Monjuvi. The words anticipate, believe, estimate, expect, intend, may, plan, predict, project, would, could, potential, possible, hope and similar expressions are intended to identify forward-looking statements, although not all forward-looking statements contain these identifying words. The forward-looking statements contained herein represent the judgment of MorphoSys as of the date of this release and involve known and unknown risks and uncertainties, which might cause the actual results, financial condition and liquidity, performance or achievements of MorphoSys, or industry results, to be materially different from any historic or future results, financial conditions and liquidity, performance or achievements expressed or implied by such forward-looking statements. In addition, even if MorphoSys' results, performance, financial condition and liquidity, and the development of the industry in which it operates are consistent with such forward-looking statements, they may not be predictive of results or developments in future periods. Among the factors that may result in differences are MorphoSys' expectations regarding risks and uncertainties related to the impact of the COVID-19 pandemic to MorphoSys business, operations, strategy, goals and anticipated milestones, including its ongoing and planned research activities, ability to conduct ongoing and planned clinical trials, clinical supply of current or future drug candidates, commercial supply of current or future approved products, and launching, marketing and selling current or future approved products, the global collaboration and license agreement for tafasitamab, the further clinical development of tafasitamab, including ongoing confirmatory trials, and MorphoSys ability to obtain and maintain requisite regulatory approvals and to enroll patients in its planned clinical trials, additional interactions with regulatory authorities and expectations regarding future regulatory filings and possible additional approvals for tafasitamab-cxix as well as the commercial performance of Monjuvi, MorphoSys' reliance on collaborations with third parties, estimating the commercial potential of its development programs and other risks indicated in the risk factors included in MorphoSys Annual Report on Form 20-F and other filings with the U.S. Securities and Exchange Commission. Given these uncertainties, the reader is advised not to place any undue reliance on such forward-looking statements. These forward-looking statements speak only as of the date of publication of this document. MorphoSys expressly disclaims any obligation to update any such forward-looking statements in this document to reflect any change in its expectations with regard thereto or any change in events, conditions or circumstances on which any such statement is based or that may affect the likelihood that actual results will differ from those set forth in the forward-looking statements, unless specifically required by law or regulation.

1 Belada D, M.D., Ph.D., et al. A Phase 1b, Open-label, Randomized Study to Assess Safety and Preliminary Efficacy of Tafasitamab (MOR208) or Tafasitamab + Lenalidomide in Addition to R-CHOP in Patients with Newly Diagnosed Diffuse Large B-Cell Lymphoma: Analysis of the Safety Run-In Phase. 62nd American Society of Hematology Annual Meeting & Exposition (ASH). Abstract #3028.

2 Maddocks KJ, M.D., et al. Long-Term Subgroup Analyses from L-MIND, a Phase 2 Study of Tafasitamab (MOR208) Combined with Lenalidomide in Patients with Relapsed or Refractory Diffuse Large B-Cell Lymphoma. 62nd American Society of Hematology Annual Meeting & Exposition (ASH). Abstract #3021.

3 Monjuvi (tafasitamab-cxix) Prescribing Information. Boston, MA, MorphoSys.

4 Sarkozy C, et al. Management of relapsed/refractory DLBCL. Best Practice Research & Clinical Haematology. 2018 31:20916. doi.org/10.1016/j.beha.2018.07.014.

5 Skrabek P, et al. Emerging therapies for the treatment of relapsed or refractory diffuse large B cell lymphoma. Current Oncology. 2019 26(4): 253265. doi.org/10.3747/co.26.5421.

6 DRG Epidemiology data.

7 Kantar Market Research (TPP testing 2018).

8 Friedberg, Jonathan W. Relapsed/Refractory Diffuse Large B-Cell Lymphoma. Hematology Am Soc Hematol Educ Program 2011; 2011:498-505. doi: 10.1182/asheducation-2011.1.498.

Continued here:
Data Evaluating Tafasitamab with and without Lenalidomide in Combination with R-CHOP in Patients with DLBCL Presented at ASH 2020 - Business Wire

Read More...

Seagen Announces Multiple ADCETRIS (brentuximab vedotin) Presentations at the 2020 ASH Annual Meeting – Business Wire

Saturday, December 12th, 2020

BOTHELL, Wash.--(BUSINESS WIRE)--Seagen Inc. (Nasdaq:SGEN) today announced multiple ADCETRIS (brentuximab vedotin) data presentations at the 62nd American Society of Hematology (ASH) Annual Meeting and Exposition, taking place virtually December 5-8, 2020. Data presentations include five-year updates from the phase 3 ECHELON-1 and ECHELON-2 clinical trials evaluating ADCETRIS plus a chemotherapy combination regimen in frontline advanced stage classical Hodgkin lymphoma (HL) and CD30-expressing frontline peripheral T-cell lymphoma (PTCL), respectively. In addition, first results were presented from an ongoing phase 2 clinical trial evaluating ADCETRIS in combination with Opdivo (nivolumab) in relapsed or refractory mediastinal gray zone lymphoma (MGZL), a rare type of non-Hodgkin lymphoma that express CD30 with no standard of care. ADCETRIS is an antibody-drug conjugate (ADC) directed to CD30, a defining marker of classical HL and expressed on the surface of several types of non-Hodgkin lymphoma, including PTCL. ADCETRIS is being evaluated globally in more than 70 corporate- and investigator-sponsored clinical trials across multiple settings in lymphoma and other indications. ADCETRIS and Opdivo are not approved alone or in combination for the treatment of relapsed or refractory MGZL.

After five years of follow-up, an important clinical milestone, both the ECHELON-1 and ECHELON-2 clinical trials demonstrate that ADCETRIS plus chemotherapy resulted in superior and durable outcomes when compared with standard chemotherapy regimens, said Roger Dansey, M.D., Chief Medical Officer at Seagen. As most relapses in Hodgkin lymphoma occur within five years of frontline treatment, the results of the ECHELON-1 study suggest that patients treated with ADCETRIS plus chemotherapy are more likely to experience long-term remissions compared to those treated with the ABVD regimen.

Brentuximab Vedotin with Chemotherapy for Patients with Previously Untreated, Stage III/IV Classical Hodgkin Lymphoma: 5-Year Update of the ECHELON-1 Study (Abstract #2973, poster presentation on Monday, December 7, 2020)

The ECHELON-1 clinical trial is evaluating ADCETRIS in combination with AVD (Adriamycin [doxorubicin], vinblastine, dacarbazine) compared to ABVD (Adriamycin [doxorubicin], bleomycin, vinblastine, dacarbazine) in patients with Stage III or IV frontline classical HL. As previously reported, the ECHELON-1 trial achieved its primary endpoint with the combination of ADCETRIS plus AVD resulting in a statistically significant improvement in modified progression-free survival (PFS) compared to the control arm of ABVD as assessed by independent review facility (IRF; hazard ratio (HR), 0.77; p=0.035). A five-year exploratory analysis was conducted to examine PFS outcomes per investigator assessment in the intent-to-treat population of 1,334 patients. Results include:

The ECHELON-2 Trial: 5-Year Results of a Randomized, Double-Blind, Phase 3 Study of Brentuximab Vedotin and CHP (A+CHP) Versus CHOP in Frontline Treatment of Patients with CD30-Positive Peripheral T-Cell Lymphoma (Abstract #1150, poster presentation on Saturday, December 5, 2020)

The ECHELON-2 clinical trial is evaluating ADCETRIS in combination with CHP (cyclophosphamide, Adriamycin [doxorubicin], prednisone) compared to CHOP (cyclophosphamide, Adriamycin [doxorubicin], vincristine, prednisone) in frontline CD30-expressing PTCL. As previously reported, the ECHELON-2 trial met its primary endpoint with the combination of ADCETRIS plus CHP resulting in a statistically significant improvement in PFS versus the control arm of CHOP per blinded independent central review (HR, 0.71; p=0.0110). A five-year post-hoc exploratory analysis was conducted to examine PFS outcome and overall survival (OS) per investigator assessment in the intent-to-treat population of 452 patients. Key findings include:

Nivolumab Combined with Brentuximab Vedotin for Relapsed/Refractory Mediastinal Gray Zone Lymphoma: Primary Efficacy and Safety Analysis of the Phase 2 CheckMate 436 Study (Abstract #2045, poster presentation on Sunday, December 6, 2020)

Data from the ongoing CheckMate 436 phase 2 clinical trial of 10 patients with relapsed or refractory MGZL who received a combination of ADCETRIS plus Opdivo treatment after autologous stem cell transplant or two or more lines of multi-agent chemotherapy if ineligible for transplant will be presented for the first time. Patients were treated once every three weeks or until disease progression or unacceptable toxicity. The median age of patients was 35 years. Key findings include:

About ADCETRIS

ADCETRIS is an ADC comprising an anti-CD30 monoclonal antibody attached by a protease-cleavable linker to a microtubule disrupting agent, monomethyl auristatin E (MMAE), utilizing Seagens proprietary technology. The ADC employs a linker system that is designed to be stable in the bloodstream but to release MMAE upon internalization into CD30-expressing cells.

ADCETRIS for injection for intravenous infusion has received FDA approval for six indications in adult patients with: (1) previously untreated systemic anaplastic large cell lymphoma (sALCL) or other CD30-expressing peripheral T-cell lymphomas (PTCL), including angioimmunoblastic T-cell lymphoma and PTCL not otherwise specified, in combination with cyclophosphamide, doxorubicin, and prednisone, (2) previously untreated Stage III or IV classical Hodgkin lymphoma (cHL), in combination with doxorubicin, vinblastine, and dacarbazine, (3) cHL at high risk of relapse or progression as post-autologous hematopoietic stem cell transplantation (auto-HSCT) consolidation, (4) cHL after failure of auto-HSCT or failure of at least two prior multi-agent chemotherapy regimens in patients who are not auto-HSCT candidates, (5) sALCL after failure of at least one prior multi-agent chemotherapy regimen, and (6) primary cutaneous anaplastic large cell lymphoma (pcALCL) or CD30-expressing mycosis fungoides (MF) who have received prior systemic therapy.

Health Canada granted ADCETRIS approval with conditions in 2013 for patients with (1) HL after failure of autologous stem cell transplant (ASCT) or after failure of at least two multi-agent chemotherapy regimens in patients who are not ASCT candidates and (2) sALCL after failure of at least one multi-agent chemotherapy regimen. Non-conditional approval was granted for (3) post-ASCT consolidation treatment of patients with HL at increased risk of relapse or progression in 2017, (4) adult patients with pcALCL or CD30-expressing MF who have received prior systemic therapy in 2018, (5) for previously untreated patients with Stage IV HL in combination with doxorubicin, vinblastine, and dacarbazine in 2019, and (6) for previously untreated adult patients with sALCL, peripheral T-cell lymphoma-not otherwise specified (PTCL-NOS) or angioimmunoblastic T-cell lymphoma (AITL), whose tumors express CD30, in combination with cyclophosphamide, doxorubicin, prednisone in 2019.

ADCETRIS received conditional marketing authorization from the European Commission in October 2012. The approved indications in Europe are: (1) for the treatment of adult patients with previously untreated CD30-positive Stage IV Hodgkin lymphoma in combination with doxorubicin, vinblastine and dacarbazine (AVD), (2) for the treatment of adult patients with CD30-positive Hodgkin lymphoma at increased risk of relapse or progression following ASCT, (3) for the treatment of adult patients with relapsed or refractory CD30-positive Hodgkin lymphoma following ASCT, or following at least two prior therapies when ASCT or multi-agent chemotherapy is not a treatment option, (4) for the treatment of adult patients with previously untreated sALCL in combination with cyclophosphamide, doxorubicin and prednisone (CHP), (5) for the treatment of adult patients with relapsed or refractory sALCL, and (6) for the treatment of adult patients with CD30-positive cutaneous T-cell lymphoma (CTCL) after at least one prior systemic therapy.

ADCETRIS has received marketing authorization by regulatory authorities in more than 70 countries for relapsed or refractory Hodgkin lymphoma and sALCL. See U.S. important safety information, including Boxed Warning, below.

Seagen and Takeda are jointly developing ADCETRIS. Under the terms of the collaboration agreement, Seagen has U.S. and Canadian commercialization rights and Takeda has rights to commercialize ADCETRIS in the rest of the world. Seagen and Takeda are funding joint development costs for ADCETRIS on a 50:50 basis, except in Japan where Takeda is solely responsible for development costs.

ADCETRIS (brentuximab vedotin) U.S. Important Safety Information

BOXED WARNING

PROGRESSIVE MULTIFOCAL LEUKOENCEPHALOPATHY (PML): JC virus infection resulting in PML and death can occur in ADCETRIS-treated patients.

Contraindication

ADCETRIS concomitant with bleomycin due to pulmonary toxicity (e.g., interstitial infiltration and/or inflammation).

Warnings and Precautions

Administer G-CSF primary prophylaxis beginning with Cycle 1 for patients who receive ADCETRIS in combination with chemotherapy for previously untreated Stage III/IV cHL or previously untreated PTCL.

Monitor complete blood counts prior to each ADCETRIS dose. Monitor more frequently for patients with Grade 3 or 4 neutropenia. Monitor patients for fever. If Grade 3 or 4 neutropenia develops, consider dose delays, reductions, discontinuation, or G-CSF prophylaxis with subsequent doses.

Most Common (20% in any study) Adverse Reactions

Peripheral neuropathy, fatigue, nausea, diarrhea, neutropenia, upper respiratory tract infection, pyrexia, constipation, vomiting, alopecia, decreased weight, abdominal pain, anemia, stomatitis, lymphopenia, and mucositis.

Drug Interactions

Concomitant use of strong CYP3A4 inhibitors or inducers has the potential to affect the exposure to monomethyl auristatin E (MMAE).

Use in Specific Populations

Moderate or severe hepatic impairment or severe renal impairment: MMAE exposure and adverse reactions are increased. Avoid use.

Advise males with female sexual partners of reproductive potential to use effective contraception during ADCETRIS treatment and for at least 6 months after the final dose of ADCETRIS.

Advise patients to report pregnancy immediately and avoid breastfeeding while receiving ADCETRIS.

Please see the full Prescribing Information, including BOXED WARNING, for ADCETRIS here.

About Seagen

Seagen is a global biotechnology company that discovers, develops and commercializes transformative cancer medicines to make a meaningful difference in peoples lives. Seagen is headquartered in the Seattle, Washington area, and has locations in California, Canada, Switzerland and the European Union. For more information on the companys marketed products and robust pipeline, visit http://www.seagen.com and follow @SeagenGlobal on Twitter.

Forward Looking Statements

Certain of the statements made in this press release are forward-looking, such as those, among others, relating to the therapeutic potential of ADCETRIS plus chemotherapy combination regimens in frontline advanced stage Hodgkin lymphoma, and frontline peripheral T-cell lymphoma, and in combination with Opdivo (nivolumab) in relapsed or refractory mediastinal gray zone lymphoma (MGZL). Actual results or developments may differ materially from those projected or implied in these forward-looking statements due to factors such as unexpected adverse events, adverse regulatory actions, the degree of utilization and adoption of an approved treatment regimen by prescribing physicians, the difficulty and uncertainty of pharmaceutical product development, negative or disappointing clinical trial results and risks related to the duration and severity of the COVID-19 pandemic. More information about the risks and uncertainties faced by Seagen is contained under the caption Risk Factors included in the Companys Quarterly Report on Form 10-Q for the quarter ended September 30, 2020 filed with the Securities and Exchange Commission. Seagen disclaims any intention or obligation to update or revise any forward-looking statements, whether as a result of new information, future events or otherwise, except as required by law.

More:
Seagen Announces Multiple ADCETRIS (brentuximab vedotin) Presentations at the 2020 ASH Annual Meeting - Business Wire

Read More...

Magenta Therapeutics and bluebird bio Announce a Phase 2 Clinical Trial Collaboration to Evaluate Magenta’s MGTA-145 for Mobilizing and Collecting…

Saturday, December 5th, 2020

CAMBRIDGE, Mass.--(BUSINESS WIRE)--Magenta Therapeutics (NASDAQ: MGTA) and bluebird bio, Inc. (NASDAQ: BLUE) today announced an exclusive clinical trial collaboration to evaluate the utility of MGTA-145, in combination with plerixafor, for mobilization and collection of stem cells in adults and adolescents with sickle cell disease (SCD). The data from this clinical trial could provide proof-of-concept for MGTA-145, in combination with plerixafor, as the preferred mobilization regimen for patients with SCD. bluebird bios experience with plerixafor as a mobilization agent in sickle cell disease aligns with Magentas combination therapy approach, utilizing MGTA-145 plus plerixafor with potential to achieve safe, rapid and reliable mobilization of sufficient quantities of high-quality stem cells to improve outcomes associated with stem cell transplantation. Under the collaboration, the stem cells will be fully characterized, and Magenta will undertake preclinical studies to evaluate the ability of these cells to be gene corrected and engrafted in mouse models. The companies will co-fund the clinical trial and Magenta will retain all rights to its product candidate.

We are excited to build upon our leading position in the field of ex-vivo gene therapy and the promising clinical data with LentiGlobin in SCD with a collaboration focused on achieving improved stem cell mobilization, said Dave Davidson, M.D., chief medical officer, bluebird bio. In this initial study, we hope to establish whether the combination of plerixafor with MGTA-145 can generate appropriate CD34+ stem cells with a single round of mobilization. If successful, we hope to evaluate this novel mobilization regimen with LentiGlobin to make another step forward in the treatment of patients with SCD.

Achieving reliable and rapid stem cell mobilization and a simplified collection process can ensure the entire patient experience is optimal with respect to therapeutic outcome. The incorporation of bluebird bios experience in this area of treatment will be immensely valuable in further developing MGTA-145 plus plerixafor to address the remaining unmet needs in gene therapy approaches for diseases like sickle cell disease, said John Davis Jr., M.D., M.P.H., M.S., Head of Research & Development and Chief Medical Officer, Magenta Therapeutics. We look forward to collaborating with bluebird bio to evaluate MGTA-145 as the preferred mobilization option for people with sickle cell disease.

SCD is a serious, progressive and debilitating genetic disease caused by a mutation in the -globin gene that leads to the production of abnormal sickle hemoglobin (HbS), causing red blood cells (RBCs) to become sickled and fragile, resulting in chronic hemolytic anemia, vasculopathy and painful vaso-occlusive events (VOEs). For adults and children living with SCD, this means unpredictable episodes of excruciating pain due to vaso-occlusion as well as other acute complicationssuch as acute chest syndrome (ACS), stroke, and infections, which can contribute to early mortality in these patients.

Currently available mobilization drugs, including granulocyte-colony stimulating factor (G-CSF), a commonly used mobilization agent administered over the course of five to seven days in other transplant settings, is not used in sickle cell disease because it can trigger vaso-occlusive crises and even death in adults and adolescents. Plerixafor is used to mobilize a patients stem cells for collection prior to transplant and while an available treatment option, multiple cycles of apheresis and collection may sometimes be required to generate sufficient stem cells for gene therapy. Magenta is developing MGTA-145, in combination with plerixafor, to be the preferred mobilization regimen for rapid and reliable mobilization and collection of hematopoietic stem cells (HSCs) to improve stem cell transplantation outcomes in multiple disease areas, including genetic diseases such as sickle cell disease, as well as blood cancers and autoimmune diseases.

About Magenta Therapeutics MGTA-145

MGTA-145, in combination with plerixafor, has demonstrated, in a recently completed Phase 1 study in healthy volunteers, it can rapidly and reliably mobilize high numbers of functional stem cells in a single day, without the need for G-CSF. MGTA-145 works in combination with plerixafor to harness a physiological mechanism of stem cell mobilization to rapidly and reliably mobilize HSCs for collection and transplant across multiple indications.

Additionally, as shown in preclinical studies, stem cells mobilized with MGTA-145 can be efficiently gene-modified and are able to engraft, potentially allowing for safer and more efficient mobilization for gene therapy approaches to treat sickle cell disease and other genetic diseases.

Magenta completed its Phase 1 trial of MGTA-145 in healthy volunteers, demonstrating MGTA-145 was well tolerated and enables same-day dosing, mobilization and simplified collection of sufficient stem cells for transplant, meeting all primary and secondary endpoints.

About bluebird bio, Inc.

bluebird bio is pioneering gene therapy with purpose. From our Cambridge, Mass., headquarters, were developing gene and cell therapies for severe genetic diseases and cancer, with the goal that people facing potentially fatal conditions with limited treatment options can live their lives fully. Beyond our labs, were working to positively disrupt the healthcare system to create access, transparency and education so that gene therapy can become available to all those who can benefit.

bluebird bio is a human company powered by human stories. Were putting our care and expertise to work across a spectrum of disorders: cerebral adrenoleukodystrophy, sickle cell disease, -thalassemia and multiple myeloma, using gene and cell therapy technologies including gene addition, and (megaTAL-enabled) gene editing.

bluebird bio has additional nests in Seattle, Wash.; Durham, N.C.; and Zug, Switzerland. For more information, visit bluebirdbio.com.

Follow bluebird bio on social media: @bluebirdbio, LinkedIn, Instagram and YouTube.

LentiGlobin and bluebird bio are trademarks of bluebird bio, Inc.

About Magenta Therapeutics

Magenta Therapeutics is a clinical-stage biotechnology company developing medicines to bring the curative power of immune system reset through stem cell transplant to more patients with autoimmune diseases, genetic diseases and blood cancers. Magenta is combining leadership in stem cell biology and biotherapeutics development with clinical and regulatory expertise, a unique business model and broad networks in the stem cell transplant world to revolutionize immune reset for more patients.

Magenta is based in Cambridge, Mass. For more information, please visit http://www.magentatx.com.

Follow Magenta on Twitter: @magentatx.

Forward-Looking Statement

This press release may contain forward-looking statements and information within the meaning of The Private Securities Litigation Reform Act of 1995 and other federal securities laws. The use of words such as may, will, could, should, expects, intends, plans, anticipates, believes, estimates, predicts, projects, seeks, endeavour, potential, continue or the negative of such words or other similar expressions can be used to identify forward-looking statements. The express or implied forward-looking statements included in this press release are only predictions and are subject to a number of risks, uncertainties and assumptions, including, without limitation risks set forth under the caption Risk Factors in Magentas Annual Report on Form 10-K filed on March 3, 2020, and in bluebird bios Annual Report on Form 10-K filed on February 18, 2020, as updated by each companys most recent Quarterly Report on Form 10-Q and its other filings with the Securities and Exchange Commission. In light of these risks, uncertainties and assumptions, the forward-looking events and circumstances discussed in this press release may not occur and actual results could differ materially and adversely from those anticipated or implied in the forward-looking statements. You should not rely upon forward-looking statements as predictions of future events. Although Magenta and bluebird bio believe that the expectations reflected in the forward-looking statements are reasonable, neither Magenta nor bluebird bio can guarantee that the future results, levels of activity, performance or events and circumstances reflected in the forward-looking statements will be achieved or occur. Moreover, except as required by law, neither Magenta or bluebird bio, nor any other person assumes responsibility for the accuracy and completeness of the forward-looking statements included in this press release. Any forward-looking statement included in this press release speaks only as of the date on which it was made. Neither Magenta nor bluebird undertake any obligation to publicly update or revise any forward-looking statement, whether as a result of new information, future events or otherwise, except as required by law.

Follow this link:
Magenta Therapeutics and bluebird bio Announce a Phase 2 Clinical Trial Collaboration to Evaluate Magenta's MGTA-145 for Mobilizing and Collecting...

Read More...

Fate Therapeutics Reports Positive Interim Data from its Phase 1 Study of FT516 in Combination with Rituximab for B-cell Lymphoma – GlobeNewswire

Saturday, December 5th, 2020

3 of 4 Patients Evaluable for Efficacy in Dose Escalation Cohorts 2 and 3 Show Objective Response, with 2 Patients Achieving Complete Response

No Observed Events of Any Grade of Cytokine Release Syndrome, Immune Effector Cell-Associated Neurotoxicity Syndrome, or Graft-vs-Host Disease

Six Doses of FT516 were Well-tolerated with No FT516-related Grade 3 or Greater Adverse Events Reported by Investigators

Management to Host Virtual Event Entitled The Power of hnCD16 Today at 4:30 PM Eastern Time

SAN DIEGO, Dec. 04, 2020 (GLOBE NEWSWIRE) -- Fate Therapeutics, Inc. (NASDAQ: FATE), a clinical-stage biopharmaceutical company dedicated to the development of programmed cellular immunotherapies for cancer and immune disorders, today announced positive interim data from the Companys dose escalation Phase 1 study of FT516 in combination with rituximab for patients with relapsed / refractory B-cell lymphoma. FT516 is the Companys universal, off-the-shelf natural killer (NK) cell product candidate derived from a clonal master induced pluripotent stem cell (iPSC) line engineered with a novel high-affinity, non-cleavable CD16 (hnCD16) Fc receptor, which is designed to maximize antibody-dependent cellular cytotoxicity (ADCC), a potent anti-tumor mechanism by which NK cells recognize, bind and kill antibody-coated cancer cells.

We are highly encouraged by these Phase 1 data, which clearly demonstrate that off-the-shelf, iPSC-derived NK cells can drive complete responses for cancer patients and that our proprietary hnCD16 Fc receptor can effectively synergize with and enhance the mechanism of action of tumor-targeted antibodies, said Scott Wolchko, President and Chief Executive Officer of Fate Therapeutics. Importantly, the safety profile of FT516 continues to suggest multiple doses of iPSC-derived NK cells can be administered in the outpatient setting, and supports potential use across multiple lines of therapy, including as part of early-line CD20-targeted monoclonal antibody regimens, for the treatment of B-cell lymphoma.

As of a November 16, 2020 data cutoff, three patients in the second dose cohort of 90 million cells per dose and one patient in the third dose cohort of 300 million cells per dose were available for assessment of safety and efficacy. All four patients were heavily pre-treated, having received at least two prior rituximab-containing regimens. Each patient received two 30-day treatment cycles, with each cycle consisting of fludarabine and cyclophosphamide lympho-conditioning followed by three once-weekly doses of FT516, IL-2 cytokine support, and rituximab.

Safety DataAll four relapsed / refractory patients were administered FT516 in an outpatient setting with no requirement for inpatient monitoring. No dose-limiting toxicities, and no cases of any grade of cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, or graft-versus-host disease, were observed. The multi-dose, two-cycle treatment regimen was well-tolerated with no FT516-related grade 3 or greater adverse events reported by investigators. In addition, no evidence of anti-product T- or B-cell mediated host-versus-product alloreactivity was detected, supporting the potential to safely administer up to six doses of FT516 in the outpatient setting without patient matching. All grade 3 or greater treatment emergent adverse events were not related to FT516 and were consistent with lympho-conditioning chemotherapy and underlying disease.

Activity DataThree of four relapsed / refractory patients achieved an objective response, including two complete responses (CR), following the second FT516 treatment cycle as assessed by PET-CT scan per Lugano 2014 criteria. A CR was achieved in one patient with diffuse large B-cell lymphoma (DLBCL) who was most recently refractory to a rituximab-containing treatment regimen, and a CR was achieved in one patient with follicular lymphoma (FL) who had previously been treated with four rituximab-containing treatment regimens. Notably, in one patient for which an interim tumor assessment showed a partial response following the first FT516 treatment cycle, the response deepened to a CR following administration of the second FT516 treatment cycle, suggesting that additional FT516 treatment cycles can confer clinical benefit.

M = million; CR = Complete Response; PR = Partial Response; PD = Progressive DiseaseAs of November 16, 2020 database entry. Data subject to cleaning and source document verification.1 Day 29 of the second FT516 treatment cycle as assessed per Lugano 2014 criteria

Dose escalation is continuing in the current dose cohort of 300 million cells per dose in combination with rituximab, and a fourth dose cohort of 900 million cells per dose in combination with rituximab is planned. The Company previously reported that two patients treated in the first dose cohort of 30 million cells per dose in combination with rituximab showed a protocol-defined response assessment of progressive disease. No events of cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, or graft-versus-host disease were observed in either patient.

About Fate Therapeutics iPSC Product PlatformThe Companys proprietary induced pluripotent stem cell (iPSC) product platform enables mass production of off-the-shelf, engineered, homogeneous cell products that can be administered with multiple doses to deliver more effective pharmacologic activity, including in combination with other cancer treatments. Human iPSCs possess the unique dual properties of unlimited self-renewal and differentiation potential into all cell types of the body. The Companys first-of-kind approach involves engineering human iPSCs in a one-time genetic modification event and selecting a single engineered iPSC for maintenance as a clonal master iPSC line. Analogous to master cell lines used to manufacture biopharmaceutical drug products such as monoclonal antibodies, clonal master iPSC lines are a renewable source for manufacturing cell therapy products which are well-defined and uniform in composition, can be mass produced at significant scale in a cost-effective manner, and can be delivered off-the-shelf for patient treatment. As a result, the Companys platform is uniquely capable of overcoming numerous limitations associated with the production of cell therapies using patient- or donor-sourced cells, which is logistically complex and expensive and is subject to batch-to-batch and cell-to-cell variability that can affect clinical safety and efficacy. Fate Therapeutics iPSC product platform is supported by an intellectual property portfolio of over 300 issued patents and 150 pending patent applications.

About FT516FT516 is an investigational, universal, off-the-shelf natural killer (NK) cell cancer immunotherapy derived from a clonal master induced pluripotent stem cell (iPSC) line engineered to express a novel high-affinity 158V, non-cleavable CD16 (hnCD16) Fc receptor, which has been modified to prevent its down-regulation and to enhance its binding to tumor-targeting antibodies. CD16 mediates antibody-dependent cellular cytotoxicity (ADCC), a potent anti-tumor mechanism by which NK cells recognize, bind and kill antibody-coated cancer cells. ADCC is dependent on NK cells maintaining stable and effective expression of CD16, which has been shown to undergo considerable down-regulation in cancer patients. In addition, CD16 occurs in two variants, 158V or 158F, that elicit high or low binding affinity, respectively, to the Fc domain of IgG1 antibodies. Scientists from the Company have shown in a peer-reviewed publication (Blood. 2020;135(6):399-410) that hnCD16 iPSC-derived NK cells, compared to peripheral blood NK cells, elicit a more durable anti-tumor response and extend survival in combination with anti-CD20 monoclonal antibodies in an in vivo xenograft mouse model of human lymphoma. Numerous clinical studies with FDA-approved tumor-targeting antibodies, including rituximab, trastuzumab and cetuximab, have demonstrated that patients homozygous for the 158V variant, which is present in only about 15% of patients, have improved clinical outcomes. FT516 is being investigated in an open-label, multi-dose Phase 1 clinical trial as a monotherapy for the treatment of acute myeloid leukemia and in combination with CD20-targeted monoclonal antibodies for the treatment of advanced B-cell lymphoma (NCT04023071). Additionally, FT516 is being investigated in an open-label, multi-dose Phase 1 clinical trial in combination with avelumab for the treatment of advanced solid tumor resistant to anti-PDL1 checkpoint inhibitor therapy (NCT04551885).

About Fate Therapeutics, Inc.Fate Therapeutics is a clinical-stage biopharmaceutical company dedicated to the development of first-in-class cellular immunotherapies for cancer and immune disorders. The Company has established a leadership position in the clinical development and manufacture of universal, off-the-shelf cell products using its proprietary induced pluripotent stem cell (iPSC) product platform. The Companys immuno-oncology product candidates include natural killer (NK) cell and T-cell cancer immunotherapies, which are designed to synergize with well-established cancer therapies, including immune checkpoint inhibitors and monoclonal antibodies, and to target tumor-associated antigens with chimeric antigen receptors (CARs). The Companys immuno-regulatory product candidates include ProTmune, a pharmacologically modulated, donor cell graft that is currently being evaluated in a Phase 2 clinical trial for the prevention of graft-versus-host disease, and a myeloid-derived suppressor cell immunotherapy for promoting immune tolerance in patients with immune disorders. Fate Therapeutics is headquartered in San Diego, CA. For more information, please visit http://www.fatetherapeutics.com.

Forward-Looking StatementsThis release contains "forward-looking statements" within the meaning of the Private Securities Litigation Reform Act of 1995 including statements regarding the safety and therapeutic potential of the Companys iPSC-derived NK cell product candidates, including FT516, its ongoing and planned clinical studies, and the expected clinical development plans for FT516. These and any other forward-looking statements in this release are based on management's current expectations of future events and are subject to a number of risks and uncertainties that could cause actual results to differ materially and adversely from those set forth in or implied by such forward-looking statements. These risks and uncertainties include, but are not limited to, the risk that results observed in studies of its product candidates, including preclinical studies and clinical trials of any of its product candidates, will not be observed in ongoing or future studies involving these product candidates, the risk that the Company may cease or delay clinical development of any of its product candidates for a variety of reasons (including requirements that may be imposed by regulatory authorities on the initiation or conduct of clinical trials or to support regulatory approval, difficulties or delays in subject enrollment in current and planned clinical trials, difficulties in manufacturing or supplying the Companys product candidates for clinical testing, and any adverse events or other negative results that may be observed during preclinical or clinical development), and the risk that its product candidates may not produce therapeutic benefits or may cause other unanticipated adverse effects. For a discussion of other risks and uncertainties, and other important factors, any of which could cause the Companys actual results to differ from those contained in the forward-looking statements, see the risks and uncertainties detailed in the Companys periodic filings with the Securities and Exchange Commission, including but not limited to the Companys most recently filed periodic report, and from time to time in the Companys press releases and other investor communications.Fate Therapeutics is providing the information in this release as of this date and does not undertake any obligation to update any forward-looking statements contained in this release as a result of new information, future events or otherwise.

Contact:Christina TartagliaStern Investor Relations, Inc.212.362.1200christina@sternir.com

Read the original post:
Fate Therapeutics Reports Positive Interim Data from its Phase 1 Study of FT516 in Combination with Rituximab for B-cell Lymphoma - GlobeNewswire

Read More...

One sneaky player prop trend to watch for – Yahoo Sport Australia

Saturday, December 5th, 2020

GlobeNewswire

Updated data from the ALLCAR study suggests AUTO1s potential for transformational activity in adult patients with r/r ALL Conference call and webcast to be held Monday, December 7, 2020at 4:00 pm ET / 9:00 pm GMTLONDON, Dec. 05, 2020 (GLOBE NEWSWIRE) -- Autolus Therapeutics plc(Nasdaq: AUTL), a clinical-stage biopharmaceutical company developing next-generation programmed T cell therapies, today announced new data highlighting progress on its AUTO1 program, the companys CAR T cell therapy being investigated in the ongoing ALLCAR Phase 1 study in relapsed / refractory adult B-Acute Lymphocytic Leukemia (ALL), during the American Society of Hematology (ASH) All-Virtual Annual Meeting, held between December 5-8, 2020.As of the November 12, 2020 data cut-off date, 20 patients with r/r ALL had received AUTO1. AUTO1 was well tolerated, with no patients experiencing Grade 3 cytokine release syndrome (CRS). Three patients (15%), all of whom had high leukemia burden (>50% blasts), experienced Grade 3 neurotoxicity (NT) that resolved swiftly with steroids.Of the 19 patients evaluable for efficacy, 16 (84%) patients achieved minimum residual disease (MRD)-negative complete response (CR) at one month. Most notably, the durability of remissions is highly encouraging. Across all treated patients, event free survival (EFS) at six and 12 months is 69% and 52% respectively. Median EFS and overall survival (OS) has not been reached at a median follow up of 16.9 months (range up to 30.5 months).The high level of sustained CRs observed with AUTO1 in adult ALL, achieved without subsequent stem cell transplant, point to a potentially transformational treatment for adult ALL, said Dr. Claire Roddie,Consultant Hematologist,UCL Cancer Institute and University College London Hospital. Despite high disease burden and despite this being a heavily pre-treated patient population on study, AUTO1 remains well tolerated. Its encouraging to also observe promising early activity and safety in indolent NHL.Adult ALL is a disease with high unmet need, whereby approximately 60% of patients relapse or are refractory to first line therapy, said Dr. Elias Jabbour, Professor of Leukemia at The University of Texas MD Anderson Cancer Center.AUTO1 is a novel CD19 CAR T candidate with an impressive clinical profile. This profile has the potential to change standard of care as a curative therapy for r/r ALL.Dr.Christian Itin, chairman and chief executive officer of Autolus, addedWe are excited about the long-term remissions observed without a need for an additional stem cell transplant. Remarkably, this outstanding result was achieved with a well-tolerated safety profile in this fragile adult ALL population. We believe the unique characteristics of AUTO1, seen in the ALLCAR study, point to the potential for AUTO1 as a standalone and transformational therapy in r/r ALL. Our Phase 1b/2 pivotal study is under way, however, with the escalating COVID-19 pandemic, enrolment projections have had to be adjusted. We now expect to enroll patients throughout 2021 with a full data set in 2022.In addition to adult ALL, the ALLCAR study was extended to patients with indolent B cell Non-Hodgkin Lymphoma (NHL) (Cohort 1), high grade B-NHL (Cohort 2) and chronic lymphocytic leukemia(CLL) (Cohort 3). As of the data cut-off date ofNovember 12, 2020, four patients in Cohort 1 had been infused with AUTO1.AUTO1 was well tolerated, with no patients experiencing Grade 2 CRS and no patients experiencing NT of any grade. All four patients achieved a Complete Metabolic Response (CMR).Investor call on Monday December 7, 2020 Management will host a conference call and webcast on Monday, December 7, 2020 at4:00 pm ET/9:00 pm GMT to discuss the ASH data. To listen to the webcast and view the accompanying slide presentation, please go to:https://www.autolus.com/investor-relations/news-and-events/events.The call may also be accessed by dialing (866) 679-5407 for U.S. and Canada callers or (409) 217-8320 for international callers. Please reference conference ID 9188389. After the conference call, a replay will be available for one week. To access the replay, please dial (855) 859-2056 for U.S. and Canada callers or (404) 537-3406 for international callers. Please reference conference ID 9188389.About Autolus Therapeutics plc Autolus is a clinical-stage biopharmaceutical company developing next-generation, programmed T cell therapies for the treatment of cancer. Using a broad suite of proprietary and modular T cell programming technologies, the company is engineering precisely targeted, controlled and highly active T cell therapies that are designed to better recognize cancer cells, break down their defense mechanisms and eliminate these cells. Autolus has a pipeline of product candidates in development for the treatment of hematological malignancies and solid tumors. For more information please visit http://www.autolus.com. About AUTO1 AUTO1 is a CD19 CAR T cell investigational therapy designed to overcome the limitations in safety - while maintaining similar levels of efficacy - compared to current CD19 CAR T cell therapies.Designed to have a fast target binding off-rate to minimize excessive activation of the programmed T cells, AUTO1 may reduce toxicity and be less prone to T cell exhaustion, which could enhance persistence and improve the ability of the programmed T cells to engage in serial killing of target cancer cells. AUTO1 is currently being evaluated in two Phase 1 studies, one in pediatric ALL and one in adult ALL. The company has also now progressed the program to a potential pivotal study, AUTO1-AL1.About AUTO1-AL1 pivotal study The AUTO1-AL1 study will enroll patients with relapsed / refractory ALL. The study will have a short Phase1b component prior to proceeding to a single arm Phase 2 study. The primary end point is overall response rate and the key secondary end points include duration of response, MRD negative CR rate and safety. The study will enroll approximately 100 patients across 30 of the leading academic and non-academic centers in the US,UKandEurope.Forward-Looking Statements This press release contains forward-looking statements within the meaning of the "safe harbor" provisions of the Private Securities Litigation Reform Act of 1995. Forward-looking statements are statements that are not historical facts, and in some cases can be identified by terms such as "may," "will," "could," "expects," "plans," "anticipates," and "believes." These statements include, but are not limited to, statements regarding the efficacy, safety and therapeutic potential of AUTO3 and the future clinical development of AUTO3 including progress, expectations as to the reporting of data, conduct and timing. Any forward-looking statements are based on management's current views and assumptions and involve risks and uncertainties that could cause actual results, performance or events to differ materially from those expressed or implied in such statements. These risks and uncertainties include, but are not limited to, the risks that Autolus preclinical or clinical programs do not advance or result in approved products on a timely or cost effective basis or at all; the results of early clinical trials are not always being predictive of future results; the cost, timing and results of clinical trials; that many product candidates do not become approved drugs on a timely or cost effective basis or at all; the ability to enroll patients in clinical trials; possible safety and efficacy concerns; and the impact of the ongoing COVID-19 pandemic on Autolus business. For a discussion of other risks and uncertainties, and other important factors, any of which could cause Autolus actual results to differ from those contained in the forward-looking statements, see the section titled "Risk Factors" in Autolus' Annual Report on Form 20-F filed with the Securities and Exchange Commission on March 3, 2020, as amended, as well as discussions of potential risks, uncertainties, and other important factors in Autolus' subsequent filings with the Securities and Exchange Commission. All information in this press release is as of the date of the release, and the company undertakes no obligation to publicly update any forward-looking statement, whether as a result of new information, future events, or otherwise, except as required by law.Contact: Lucinda Crabtree, PhD Vice President, Investor Relations and Corporate Communications +44 (0)7587 372 619 l.crabtree@autolus.comJulia Wilson +44 (0) 7818 430877 j.wilson@autolus.comSusan A. Noonan S.A. Noonan Communications +1-212-966-3650 susan@sanoonan.com

More here:
One sneaky player prop trend to watch for - Yahoo Sport Australia

Read More...

IN8bio announces first-in-human Phase 1 trial Update from The University of Kansas Cancer Center using INB-100, IN8bio’s Gamma Delta T-cell product…

Friday, December 4th, 2020

NEW YORK, Dec. 03, 2020 (GLOBE NEWSWIRE) -- IN8bio, Inc., a clinical-stage biotechnology company focused on developing innovative allogeneic, autologous and genetically modified gamma-delta T cell therapies for the treatment of cancers (IN8bio or the Company), today announced an upcoming presentation that provides an update of the ongoing Phase I clinical trial of their product candidate INB-100 at the 62nd American Society of Hematology Annual Meeting & Exposition (ASH), which will take place virtually from December 5 to 8, 2020. INB-100 is designed for the treatment of patients with leukemia undergoing hematopoietic stem cell transplantation with haploidentical donors.

The poster and accompanying narrated slide presentation is titled, First-in-Human Phase I Trial of Adoptive Immunotherapy with Ex Vivo Expanded and Activated gamma delta T-Cells Following Haploidentical Bone Marrow Transplantation and Post-BMT Cyclophosphamide and reviews the study design and provides a brief update on enrollment and patient status.

The company reported that, as of abstract submission, three female subjects with acute leukemia had been enrolled in the INB-100 Phase 1 trial, of whom two had been dosed, and that no treatment-related adverse events had been recorded. The trial is continuing to enroll and treat patients. The abstract for the presentation can be found at https://ash.confex.com/ash/2020/webprogram/Paper142876.html.

The poster and slide presentation are jointly authored by the scientific and physician investigators from IN8bio and The University of Kansas Cancer Center (KU Cancer Center), and will be presented by the studys Principal Investigator, Dr. Joseph McGuirk, Schutte-Speas Professor of Hematology-Oncology, Division Director of Hematological Malignancies and Cellular Therapeutics and Medical Director, Blood and Marrow Transplant at KU Cancer Center.

This preliminary data report from KU Cancer Center with our allogeneic product candidate, INB-100, demonstrates the absence of significant GvHD in these initial patients, said William Ho, Chief Executive Officer of IN8bio. This suggests that gamma delta T-cells delivered as an off-the-shelf allogeneic cell therapy may be well tolerated and have significant potential to treat patients with serious and life-threatening cancers.

Dr. McGuirk, commented, Potentially curative stem cell transplants using partially matched donors -- called haploidentical transplants have greatly expanded access to stem cell transplantation. The infusion of donor-derived gamma delta T-cells from the stem cell donor, offers the hope of diminishing this risk of relapse and curing more patients.

About IN8bioIN8bio is a clinical-stage biotechnology company focused on developing novel therapies for the treatment of cancers, including solid tumors, by employing allogeneic, autologous and genetically modified gamma-delta T cells. IN8bios technology incorporates drug-resistant immunotherapy (DRI), which has been shown in preclinical studies to function in combination with therapeutic levels of chemotherapy. IN8bio is currently conducting two investigator-initiated Phase 1 clinical trials for its lead gamma-delta T cell product candidates: INB-200 for the treatment of newly diagnosed glioblastoma, which is a difficult to treat brain tumor that progresses rapidly, and INB-100 for the treatment of patients with acute leukemia undergoing hematopoietic stem cell transplantation. For more information about the Company and its programs, visit http://www.IN8bio.com.

Forward Looking StatementsCertain statements herein concerning the Companys future expectations, plans and prospects, including without limitation, the Companys current expectations regarding the curative potential of its product candidates, constitute forward-looking statements. The use of words such as may, might, will, should, expect, plan, anticipate, believe, estimate, project, intend, future, potential, or continue, the negative of these and other similar expressions are intended to identify such forward looking statements. Such statements, based as they are on the current expectations of management, inherently involve numerous risks and uncertainties, known and unknown, many of which are beyond the Companys control. Consequently, actual future results may differ materially from the anticipated results expressed in such statements. Specific risks which could cause actual results to differ materially from the Companys current expectations include: scientific, regulatory and technical developments; failure to demonstrate safety, tolerability and efficacy; final and quality controlled verification of data and the related analyses; expense and uncertainty of obtaining regulatory approval, including from the U.S. Food and Drug Administration; and the Companys reliance on third parties, including licensors and clinical research organizations. Do not place undue reliance on any forward-looking statements included herein, which speak only as of the date hereof and which the Company is under no obligation to update or revise as a result of any event, circumstances or otherwise, unless required by applicable law.

Contact:IN8bio, Inc.Kate Rochlin, Ph.D.+1 646.933.5605info@IN8bio.com

Investor Contact:Julia Balanova+ 1 646.378.2936jbalanova@soleburytrout.com

Media Contact:Ryo Imai / Robert Flamm, Ph.D.Burns McClellan, Inc.212-213-0006 ext. 315 / 364Rimai@burnsmc.com/rflamm@burnsmc.com

Read the original:
IN8bio announces first-in-human Phase 1 trial Update from The University of Kansas Cancer Center using INB-100, IN8bio's Gamma Delta T-cell product...

Read More...

Possible Role for Comprehensive Molecular ProfilingBased Treatment Selection in Newly Diagnosed AML, Study Suggests – Cancer Therapy Advisor

Friday, December 4th, 2020

The feasibility of a precision medicinebased approach was demonstrated for patients with newly diagnosed acute myeloid leukemia (AML), according to findings from a phase 1/2 clinical trial reported in Nature Medicine.

The current standard of care for the treatment of patients diagnosed with AML involves prompt initiation of intensive induction chemotherapy, such as 7 days of standard-dose cytarabine and 3 days of daunorubicin, or administration of a hypomethylating agent for those deemed unable to tolerate standard induction therapy, to prevent rapid progression of disease in this predominantly older patient population.

Hence, time for comprehensive molecular characterization of the disease is not built into typical treatment protocols for patients with newly diagnosed AML. However, long-term outcomes of patients with newly diagnosed AML treated with intensive chemotherapy without autologous hematopoietic stem cell transplantation have been shown to be poor, and hypomethylating agents are not a curative approach in the setting of AML.

This nonrandomized, open-label, multicenter, umbrella protocol study sponsored by the Leukemia & Lymphoma Society (BEAT AML Master Trial; ClinicalTrials.gov Identifier: NCT03013998) enrolled adult patients with suspected AML prior to the administration of frontline treatment.

During a 7-day period prior to treatment assignment, bone marrow biopsy specimens of eligible patients were subjected to cytogenetic analysis, comprehensive molecular profiling using next-generation sequencing, and a FLT3-ITD ratio testing. On the basis of these results, patients with a dominant AML clone characterized by an actionable alteration were assigned to 1 of multiple molecularly defined substudy treatment arms, whereas those without evidence of such an alteration were assigned to the marker-negative subgroup.

In describing the purpose of this study, the investigators stated that they collaboratively implemented a new prospective clinical trial approach aimed at facilitating frontline treatment assignments to specific genomic-defined AML subtypes.

Of the 395 eligible patients, approximately 95% were assigned to treatment within 7 days of bone marrow biopsy collection. Of note, only 26 of these patients exhibited evidence of rapid disease progression necessitating initiation of therapy during the 7-day testing window.

The most common mutational drivers identified were DNMT3A (22.7%), TET2 (19.6%), TP53 (19.1%), ASXL1 (19.1%) and SRSF2 (18.4%).

Regarding molecularly based treatment assignment, the study authors commented that these data show that there were few co-occurring dominant mutations that could have been used for an alternative therapeutic assignment.

Only 224 (56.7%) of patients agreed to receive treatment according to their assigned BEAT AML substudy treatment arm, with 103, 28, and 38 patients selecting standard-of-care treatment, alternative investigational therapy, and palliative care, respectively.

Patients were encouraged to select an alternative therapy (alternative investigational therapy, [standard of care] or palliative care) if the patient with their health-care providers deemed this a better option, the study investigators noted.

A key finding from this study was the 30-day mortality of patients starting at initial study enrollment was 3.7% for patients enrolled on the BEAT AML trial protocol and 20.4% for those who choose to receive standard-of-care therapy.

Furthermore, rates of 1-year overall survival were 54.7%, 27.6%, 11%, and 57.4% for patients treated on the BEAT AML protocol, or with standard-of-care therapy, palliative care, and alternative investigational therapy, respectively.

However, the study investigators noted that while our study demonstrates the feasibility of precise molecular treatment assignment in older adults with AML, it does not clearly differentiate the benefit of treatment assignment based on a molecular target from better outcome that occurs simply from enrolling on a clinical trial.

They also emphasized that this approach requires a detailed team-coordinated effort by investigators, patients and caregivers, genomic laboratories, cytogenetic laboratories and a central treatment assignment team.

In their concluding remarks, the researchers commented that randomization of specific large genomic groups to targeted therapy versus [standard of care] or, in less common genomic groups, comparison of treatment with targeted therapy to either real-world data or synthetic controls, will be required to determine the comparative effectiveness of a precision medicine-based approach vs standard-of-care therapy in patients with newly diagnosed AML.

Reference

Burd A, Levine RL, Ruppert AS, et al. Precision medicine treatment in acute myeloid leukemia using prospective genomic profiling: feasibility and preliminary efficacy of the Beat AML Master Trial. Nat Med. Published online October 26, 2020. doi:10.1038/s41591-020-1089-8

See the article here:
Possible Role for Comprehensive Molecular ProfilingBased Treatment Selection in Newly Diagnosed AML, Study Suggests - Cancer Therapy Advisor

Read More...

Initiation of First UniCAR Cellular Immunotherapy Study in Advanced Solid Tumors – BioSpace

Friday, December 4th, 2020

DRESDEN, Germany, Dec. 3, 2020 /PRNewswire/ -- GEMoaB, a biopharmaceutical company focused on the development of next-generation immunotherapies for hard-to-treat cancers, today announced that it has apheresed the first patient in a Phase IA study with UniCAR-T-PSMA, the lead solid tumor product candidate from its proprietary UniCAR cellular immunotherapy platform. UniCAR-T-PSMA is investigated in late-stage, relapsed/refractory solid tumors expressing the PSMA antigen. The UniCAR platform has been designed to ensure excellent control over the universal CAR-T effector cell through a rapidly switchable on/off capability. This is combined with high flexibility to effectively target tumor antigens of choice by re-directing and activating UniCAR-T cells through soluble adapters termed Targeting Modules (TMs).

"The unique ability to rapidly switch on and off the UniCAR-T effector cells and thereby tightly control their activity as we have now clinically validated in our ongoing UniCAR-T-CD123 study in AML may help to overcome many of the limitations that conventional CAR-T therapies face when targeting less differentially expressed antigens, especially in solid tumors", said Prof. Dr. Gerhard Ehninger, GEMoaB's co-founder and Chief Medical Officer. "Our first UniCAR clinical study in solid tumors is of utmost importance for GEMoaB. We believe that the PSMA antigen is a great initial target as it is not only expressed on the tumor surface but also on the tumor neo-vasculature, allowing for a double attack of the malignant cells by UniCAR-T cells."

The Phase IA study includes patients with late-stage PSMA-positive relapsed/refractory solid tumors such as Castrate Resistant Prostate Cancer (CRPC), Non-small Cell Lung Cancer (NSCLC) or Triple Negative Breast Cancer (TNBC). It will examine the feasibility, safety and potential efficacy of the combined application of a single dose of UniCAR-T and the continuous infusion of the PSMA-specific TMpPSMA.

According to Prof. Dr. Ralf Bargou, Head of Comprehensive Cancer Center Mainfranken at the University Hospital Wrzburg and coordinating investigator of this trial, the study could be an important step in the ongoing intensive research efforts to establish cellular immunotherapies as a key therapeutic pillar to improve patient outcomes in hard-to-treat solid tumor cancers. "At our National Cancer Center in Wrzburg we are focusing a significant amount of our ongoing research and clinical efforts on developing breakthrough immunologic treatments of solid tumors together with our partners", said Prof. Bargou. "PSMA is a very promising target expressed in multiple late-stage cancers that do not sufficiently benefit from currently existing therapies and the UniCAR platform provides many features to finally obtain meaningful safety and efficacy results for this innovative treatment modality. We are very much looking forward working closely with the GEMoaB team on this important study."

About the UniCAR-T-PSMA Study

This first-in-human phase I study is an open-label, non-randomized, dose-finding study designed to evaluate the safety and activity of UniCAR-T-PSMA in up to 16 patients with advanced relapsed/refractory, PSMA-positive solid tumors such as CRPC, NSCLC or TNBC. Its purpose is to determine the maximum tolerated dose (MTD), dose limiting toxicities (DLT) as well as the recommended Phase II dose for the combined application of a single dose of UniCAR-T and the continuous infusion of TMpPSMA over 25 days. The study will also investigate response rates, persistence of UniCAR-T cells over time as well as the ability to rapidly switch UniCAR-T cells on and off in case of side effects through stopping the TM infusion. The study will take place at selected Phase I and CAR-T experienced University centers in Germany. It is supported by a grant from the European Regional Development Fund (ERDF) provided through Saxony's Development Bank (SAB). To learn more about the trial, please visit clinicaltrials.gov.

About UniCAR

GEMoaB is developing a rapidly switchable universal CAR-T platform, UniCAR, to improve the therapeutic window and increase efficacy and safety of CAR-T cell therapies in challenging cancers, including acute leukemias and solid tumors. Conventional CAR-T cells depend on the presence and direct binding of cancer antigens for activation and proliferation. An inherent key feature of the UniCAR platform is a rapidly switchable on/off mechanism (less than 4 hours after interruption of TM supply) enabled by the short pharmacokinetic half-life and fast internalization of soluble adaptors termed TMs. These TMs provide the antigen-specificity to activate UniCAR gene-modified T-cells (UniCAR-T) and consist of a highly flexible antigen binding moiety, linked to a small peptide motif recognized by UniCAR-T.

About GEMoaB

GEMoaB is a privately-owned, clinical-stage biopharmaceutical company that is aiming to become a fully integrated biopharmaceutical company. By advancing its proprietary UniCAR, RevCAR and ATAC platforms, the company will discover, develop, manufacture and commercialize next-generation immunotherapies for the treatment of cancer patients with a high unmet medical need.

GEMoaB has a broad pipeline of product candidates in pre-clinical and clinical development for the treatment of hematological malignancies as well as solid tumors. Its clinical stage assets GEM333, an Affinity-Tailored Adaptor for T-Cells (ATAC) with binding specificity to CD33 in relapsed/refractory AML, and GEM3PSCA, an ATAC with binding specificity to PSCA for the treatment of castrate-resistant metastatic prostate cancer and other PSCA expressing late stage solid tumors, are currently investigated in Phase I studies and globally partnered with Bristol-Myers Squibb. A Phase IA dose-finding study of the first UniCAR asset in hematological malignancies, UniCAR-T-CD123 for treatment of relapsed/refractory AML, is currently recruiting patients.

Manufacturing expertise, capability and capacity are key for developing cellular immunotherapies for cancer patients. GEMoaB has established a preferred partnership with its sister company Cellex in Cologne, a world leader in manufacturing hematopoietic blood stem cell products and a leading European CMO for CAR-T cells, co-operating in that area with several large biotech companies.

More information can be found at http://www.gemoab.com.

For further information please contact

Jana Fiebigerj.fiebiger@gemoab.com; Tel.: +49 351 4466-45012

Investor Contact

Michael Pehlm.pehl@gemoab.com; Tel.: +49 351 4466-45030

Forward-looking Statements This announcement includes forward-looking statements that involve risks, uncertainties and other factors, many of which are outside of our control, that could cause actual results to differ materially from the results and matters discussed in the forward looking statements. Forward looking statements include statements concerning our plans, goals, future events and or other information that is not historical information. The Company does not assume any liability whatsoever for forward-looking statements. The Company assumes that potential partners will perform and rely on their own independent analyses as the case may be. The Company will be under no obligation to update the Information.

Logo- https://mma.prnewswire.com/media/947610/GEMoaB_Logo.jpg

View original content:http://www.prnewswire.com/news-releases/initiation-of-first-unicar-cellular-immunotherapy-study-in-advanced-solid-tumors-301185515.html

SOURCE GEMoaB

See the rest here:
Initiation of First UniCAR Cellular Immunotherapy Study in Advanced Solid Tumors - BioSpace

Read More...

Transcriptome and regulatory maps of decidua-derived stromal cells inform gene discovery in preterm birth – Science Advances

Friday, December 4th, 2020

Abstract

While a genetic component of preterm birth (PTB) has long been recognized and recently mapped by genome-wide association studies (GWASs), the molecular determinants underlying PTB remain elusive. This stems in part from an incomplete availability of functional genomic annotations in human cell types relevant to pregnancy and PTB. We generated transcriptome (RNA-seq), epigenome (ChIP-seq of H3K27ac, H3K4me1, and H3K4me3 histone modifications), open chromatin (ATAC-seq), and chromatin interaction (promoter capture Hi-C) annotations of cultured primary decidua-derived mesenchymal stromal/stem cells and in vitro differentiated decidual stromal cells and developed a computational framework to integrate these functional annotations with results from a GWAS of gestational duration in 56,384 women. Using these resources, we uncovered additional loci associated with gestational duration and target genes of associated loci. Our strategy illustrates how functional annotations in pregnancy-relevant cell types aid in the experimental follow-up of GWAS for PTB and, likely, other pregnancy-related conditions.

Spontaneous preterm birth (PTB), defined as spontaneous labor and birth before 37 weeks of gestation, is associated with considerable infant mortality and morbidity, as well as long-term health consequences into adulthood (1). A genetic component to PTB has long been recognized, but the significant role of environmental factors and the etiologic heterogeneity of birth before 37 weeks (24) have made it challenging to discover genetic associations and causal genes. For example, recent genome-wide association studies (GWASs) of gestational duration in 43,568 women (3331 with a preterm delivery) (5) and in 84,689 infants (4775 born preterm) (6) reported six and one genome-wide significant associations, respectively, with gestational duration considered as a continuous variable. Three loci were also associated with PTB (defined as a categorical variable of birth) in the maternal GWAS (5), but no loci were associated with PTB in the infant GWAS (6). These studies highlight the challenges of such complex and multifactorial phenotypes and the need for additional approaches to facilitate discovery of genes contributing to gestational duration and PTB.

Integrating GWAS that results with genomic and epigenomic annotations is a promising approach for assigning function to variants discovered by GWAS, as well as for identifying additional associations that do not reach stringent genome-wide significance threshold (7, 8). While large consortia [e.g., ENCODE (Encyclopedia of DNA Elements) (9), GTEx (Genotype-Tissue Expression Project) (10), and Roadmap Epigenomics (11)] have generated annotations of putative functional elements and genetic variants for many human cell types and tissues, there is a remarkable absence in these databases for the cell types and tissues that are relevant to pregnancy in general and to PTB in particular. Because the regulation of transcription has strong cell typespecific components and because annotations in disease-relevant tissues or cells tend to be most enriched among GWAS signals for those specific diseases (10, 12), follow-up studies of GWASs of pregnancy-associated conditions have been disadvantaged compared to most other complex diseases due to the paucity of functional annotations in cells relevant to pregnancy. To fill this gap in knowledge, we characterized the transcriptional and chromatin landscapes of cultured mesenchymal stromal/stem cells (MSCs) collected from human placental membranes and decidualized MSCs, also known as decidual stromal cells (DSCs). These cells play critical roles in promoting successful pregnancy, interfacing with fetal cells throughout pregnancy, and the timing of birth (13, 14). We then built a computational framework that integrated these decidua-derived stromal cell annotations with the results of a large GWAS of gestational duration to facilitate discovery of PTB genes.

This integrated analysis revealed a significant enrichment of heritability estimates for gestational duration in decidua-derived stromal cell genomic regions marked by open chromatin or histone marks. Leveraging those functional annotations in a Bayesian statistical framework, we discovered additional loci associated with gestational duration and improved fine mapping in regions associated with gestational duration. Last, using promoter capture Hi-C (pcHi-C), we linked functionally annotated gestational age-associated variants to their putative target genes. More generally, these functional annotations should prove a valuable resource for studying other pregnancy-related conditions, such as preeclampsia and recurrent miscarriage, as well as conditions associated with endometrial dysfunction, such as endometriosis and infertility.

Decidualization is the process of transformation of endometrial MSCs into DSCs that is induced by progesterone production that begins during the luteal phase of the menstrual cycle and then increases throughout pregnancy when successful implantation occurs [reviewed in (15)]. Using progesterone and estrogen or cyclic adenosine 5-monophosphate (cAMP) to induce decidualization of MSCs in culture has been used in cells derived from endometrial biopsies in nonpregnant women to characterize their transcriptomes and epigenomes and to identify genes and molecular pathways involved in this process (1621).

Because obtaining endometrial cells in nonpregnant women through biopsies requires an invasive procedure that carries some risk and MSCs can also be obtained from human placentas (2224), we isolated these cells from the decidua parietalis of three women who had delivered at term and established one primary MSC line from each to model the process of decidualization (see Materials and Methods). Briefly, cells were treated with medroxyprogesterone acetate (MPA) and cAMP for 48 hours, and a paired set of untreated samples was cultured in parallel for 48 hours. Three replicates of treated/untreated sets of each cell line were studied to assess experimental variability in the two conditions. Each of the 18 samples (3 individual lines 3 replicates 2 conditions) were assayed to generate transcriptome [RNA sequencing (RNA-seq)], open chromatin [assay for transposase-accessible chromatin sequencing (ATAC-seq)], and histone modification [chromatin immunoprecipitation sequencing (ChIP-seq)] maps. A summary of those data is shown in table S1, and a representative example of the full set of annotations for one primary cell line is shown in Fig. 1. The number of reads generated for each sample in each condition and other descriptive data are provided in data file S1.

Each histone modification and RNA-seq track shows read counts per base pair for each experiment. The pcHi-C signal track shows the number of reads per MboI restriction fragment. Arcs in the pcHi-C interactions track show significant interactions between the promoter of the PRL gene and putative distal regulatory elements identified with pcHi-C. Pooled data (three replicates) for one cell line are shown for untreated cells (MSCs, in green) and decidualized cells (DSCs, in purple). pcHi-C data were generated in a fourth cell line that was decidualized.

Analysis of the RNA-seq data using DESeq2 (25) revealed 1135 differentially expressed genes after decidualization (table S1). Genes with decreased expression after 48 hours of treatment were highly enriched for cell cycle genes (data file S2), consistent with observations from endometrial biopsies from nonpregnant women that decidualization is associated with cell cycle arrest (19, 26). Genes with increased expression after treatment were enriched for insulin-related terms, also consistent with previous results from endometrial biopsies (26), and for glucose metabolism (18).

To identify putative regulatory elements in MSCs and DSCs, we assayed H3K27ac, H3K4me1, and H3K4me3 histone modifications, which are markers of active enhancers, poised enhancers, and active promoters, respectively [reviewed in (27)]. We also used ATAC-seq to identify open chromatin regions to complement ChIP-seq data. To identify regulatory regions that might be altered in response to, and potentially regulate decidualization, we compared read counts of ATAC-seq and ChIP-seq peaks in untreated and decidualized cells, revealing tens of thousands of regions that differed between untreated and treated samples (table S1). Most of the differential peaks were marked with H3K27Ac and H3K4me1, indicating that the epigenetic changes underlying alterations in gene expression during decidualization predominantly occur in distant regulatory elements, such as enhancers.

We observed a moderate degree of overlap between the differential peaks across ATAC-seq and ChIP-seq data, with the two enhancer marks, H3K27ac and H3K4me1, showing the most overlap (Fig. 2A). In addition, putative regulatory regions that showed chromatin changes in response to decidualization were associated with genes whose expression also changed in response to decidualization (Fig. 2B). Regulatory regions with increased read counts clustered around genes that were more highly expressed after decidualization, indicating increased chromatin accessibility or activation of enhancers of those genes. Conversely, genes that were more lowly expressed after decidualization were enriched for enhancers that became less accessible or active. These observations indicate that the differential peaks of open chromatin and histone marks observed after decidualization correspond to regulatory elements that become more or less active, resulting in correlated gene expression changes of the nearby genes.

(A) Plot showing the overlap between the different histone modifications and ATAC-seq maps (intersection between annotations). Peaks were assigned to 100-bp bins to avoid ambiguity in overlap due to different peak borders. Black circles indicate overlap with other annotations; light gray circles indicate that the annotation does not overlap others. (B) Each data point shows the ratio between the number of increased/decreased differential peaks nearby genes that increase expression after decidualization (blue, positive log ratios; upper half of the figure) or decrease expression after decidualization (orange, negative log ratios; lower half of the figure). Genes that were more highly expressed in decidualized cells were flanked by a higher number of ChIP-seq and ATAC-seq peaks that displayed increased read counts in decidualized samples compared to peaks that displayed decreased read counts (top inset). Genes that were down-regulated in decidualized cells showed the opposite trend (bottom inset). All enrichments: P < 1025. (C) DNA binding motifs of transcription factors relevant in decidualization are enriched in peaks that change following decidualization treatment. Motifs are color-coded by similarity. (D) Colocalization of PGR, FOSL2, FOXO1, GATA2, and NR2F2 with ATAC-seq and ChIP-seq peaks. Transcription factor binding sites co-occur with ATAC-seq and ChIP-seq peaks in both untreated (green) and decidualized (purple) cells more often than with random peaks. Enrichment of the co-occurrences of PGR, FOXO1, GATA2, and NR2F2 are higher when co-occurring with peaks that have increased read counts (navy blue) and lower with peaks that have decreased read counts (orange) in decidualized compared to untreated cells. Enrichment of co-occurrences with peak sets was calculated as the fold difference between the number of transcription factor peaks overlapping with ATAC-seq/ChIP-seq peaks and with a random set of peaks (see Materials and Methods).

Previous work identified transcription factors that play critical roles in decidualized stromal cells (2832). Several of the DNA binding motifs that were enriched in peaks with increased or decreased read counts in our data correspond to transcription factors previously implicated in decidualization (Fig. 2C), such as CAAT-enhancer binding protein (CEBP) (33), progesterone receptor (PGR) (28) that shares the same motif with androgen response element, and glucocorticoid receptor, FOSL2 (Fos-related antigen 2) (28), that shares the same motif with Fra1 (Fos-related antigen 1), Atf3 (Activating transcription factor 3), and BATF (basic leucine zipper ATF-like transcription factor), and TEA (transcriptional enhancer factor) domain transcription factors (21, 34). Whereas CEBP and PGR were exclusively enriched in peaks with increased read counts in decidualized cells, the FOSL2 motif was present in peaks that both changed positively and negatively in decidualized cells.

To better understand the role of these transcription factors in decidualization, we obtained publicly available ChIP-seq data for PGR (28) and FOSL2 (28) from endometrial biopsies and analyzed the colocalization of their binding locations with the putative regulatory elements identified by ATAC-seq and ChIP-seq identified in our study (Fig. 2B). We additionally analyzed FOXO1 (Forkhead box O1) (29), NR2F2 (nuclear receptor subfamily 2 group F member 2) (30), and GATA2 (GATA binding protein 2) (31) ChIP-seq data because these transcription factors have also been implicated in decidualization (2931). With the exception of FOSL2, the colocalization enrichments of PGR, FOXO1, GATA2, and NR2F2 with ATAC-seq and ChIP-seq peaks were higher (9 to 16 folds) among peaks that were increased in decidualized cells (more open chromatin or increased histone modification levels) compared to all peaks (7.5 to 12.8 folds) and to peaks that decreased in decidualized cells (2 to 5 folds). This observation supports the notion that these transcription factors are involved in regulation of decidualization (2831, 35). Although FOSL2 has been reported as a positive coregulator of PGR (28), the presence of FOSL2 motifs in peaks that both increased and decreased in decidualized cells (Fig. 2C) and the lack of difference in the colocalization enrichment between these two sets of peaks (Fig. 2D) suggests that FOSL2 may have a dual role in decidualization.

Together, our results support a model of decidualization that involves changes in the regulatory landscape during the differentiation of MSCs into DSCs, including alterations in chromatin accessibility and in the activation levels of distant regulatory elements, accompanied by the differential binding of key transcription factors, resulting in increases or decreases in gene expression.

As shown in Fig. 2B, the surrounding regions of differentially expressed genes were enriched for differential ChIP-seq and ATAC-seq peaks that changed in the same direction as the genes in decidualized samples. Accordingly, when we paired differential peaks with the nearest expressed gene as its putative gene target, we observed that these pairs were more likely to have matching directions of change (i.e., both the peak and the gene have increased or decreased read counts in decidualized samples) than nonmatching directions when compared with pairs that were assigned randomly (Fig. 3A).

(A) Randomly assigning a gene to a peak (see Materials and Methods) resulted in fewer peaks that matched the direction of change with that of differentially expressed genes than when using pcHi-C interactions or the nearest gene to pair peaks to genes. (B) The FOXO1 gene is more highly expressed in decidualized samples (fourfold increase, P = 7 1022) and its promoter physically interacts (red arcs) with distal regulatory elements (yellow highlights) that show increased activation in decidualized samples. The nearest expressed gene to these differential peaks is COG6.

In many cases, however, the target gene for a regulatory element is not the nearest gene (36), and therefore, information about distal chromatin interactions can be useful in prioritizing candidate gene targets of variants identified in GWAS. To this end, we generated a pcHi-C map of a decidualized cell line, thus enriching for the identification of long-range chromatin interactions between promoters and distant regulatory elements (3739). We identified a total of 161,337 interactions, of which 53,211 were between promoters and distal regions of accessible chromatin assayed by ATAC-seq and ChIP-seq, suggestive of their regulatory role. We used the significant interactions between promoters and distal regions that we identified to pair differential peaks with putative target genes. As shown in Fig. 3A, using pcHi-C interactions as a pairing method resulted in enhanced identification of differential peak/differential target gene pairs that have matching directions of change compared to random assignment of gene-target pairs.

Whereas assigning peaks to the nearest expressed gene also led to enhanced assignment of differential peaks to target genes with matching directions of change (Fig. 3A), pcHi-C was helpful in identifying less obvious target genes, as shown in Fig. 3B. In this example, several pcHi-C interactions link distal regulatory elements up to 847 kb away that became more active in decidualized cells to the promoter of a gene (FOXO1) that was up-regulated in decidualized cells and is known to be involved in decidualization (32). The nearest expressed gene method assigned those differential peaks to COG6, a gene that does not change expression in decidualized samples and is therefore a less likely target.

In conclusion, by combining pcHi-C interactions with the epigenome maps and transcriptome data, we were able to identify genes and putative regulatory elements that respond to, or regulate, the decidualization process. We next used these functional genomic maps and datasets to fine map GWAS loci for gestational duration and identify new candidate genes with a potential role in PTB.

To identify candidate genes that may play a role in gestational duration and PTB, we used summary data from a GWAS of gestational duration based on a meta-analysis of a 23andMe GWAS (n = 42,121) (5) and the results from six European datasets (n = 14,263). A detailed description of the GWAS is in the Supplementary Materials and figs. S1 and S2. After filtering for single-nucleotide polymorphism (SNPs) that are present in the 1000 Genomes Project data and minor allele frequency of >0.01, we identified SNPs at six autosomal loci, defined as approximately independent blocks by LDetect (40), that were associated with gestational duration at genome-wide significance of P < 5 108 (table S2). We then created a computational pipeline to assess enrichment of GWAS signals in functional annotations that we generated in untreated (MSCs) and decidualized (DSCs) stromal cells to fine map GWAS loci and discover candidate causal genes and to potentially provide support for additional loci that did not reach genome-wide significance in the GWAS (Fig. 4A). Each step of this procedure is explained below and described in details in Materials and Methods.

(A) Computational pipeline for analyzing GWAS of gestation duration. Yellow boxes (input data): GWAS summary statistics and functional annotations from endometrial stromal cells (in both untreated and decidualized cells). Green boxes: Stages of statistical analysis (see Materials and Methods). (B) Stratified LDSC heritability analysis of GWAS of gestational duration using functional annotations. Left: Fold enrichment of heritability in each annotation. Dashed line shows values at 1, i.e., no enrichment. Center: Proportion of heritability explained by each annotation. Right: Proportion of SNPs across the genome that fall within an annotation. For each annotation, enrichment (left) is the ratio of h2 proportion (center) divided by the SNP proportion (right). Error bars represent 95% confidence intervals.

We first used stratified linkage disequilibrium (LD) score regression (S-LDSC) (41) to assess enrichment of GWAS signals in functional annotations in endometrial stromal cells. S-LDSC takes as input GWAS summary statistics across the genome and functional annotations of SNPs, e.g., whether an SNP is in ATAC-seq peak, and returns as output heritability enrichment of each annotation. S-LDSC is a commonly used tool for estimating the proportion of heritability of complex phenotypes that is explained by variants in certain functional annotations. The heritability enrichment is defined by the proportion of heritability explained by annotations divided by the expected proportion, which is the percent of SNPs genome wide that are in these functional annotations. To account for possible systematic bias in this analysis, i.e., SNPs within annotations of interest may differ from background SNPs in systematic ways such as their LD structure and epigenomic properties, we included a range of baseline annotations (default S-LDSC setting), including LD-related annotations, deoxyribonuclease (DNase) hypersensitivity, enhancer annotation, H3K27ac, H3K4me1, and other histone marks (the union across cell types). Thus, if an annotation is shared by many cell types, then it would not show the enrichment in S-LDSC analysis (see Materials and Methods).

Using S-LDSC, we found 5- to 10-fold enrichments of GWAS heritability for gestational duration in our functional annotations compared to the baseline model of S-LDSC (Fig. 4). The enrichment of enhancer marks H3K27ac and H3K4me1 was higher in decidualized than in untreated cells, but the opposite pattern was observed for the promoter mark H3K4me3, which was more enriched in untreated (MSCs) than in decidualized (DSCs) cells. These findings are consistent with previous observations that enhancers are often more dynamic and condition- or tissue-specific than promoters (10). We observed weaker heritability enrichments of open chromatin regions defined by ATAC-seq and of interaction regions in pcHi-C. However, because we performed joint analysis of all annotations together, the enrichment of one annotation (e.g., ATAC-seq peaks) will be reduced if the enrichment is partially explained by other, overlapping annotations (e.g., H3K27ac). Although the promoter mark H3K4me3 in untreated cells showed the highest enrichment, the annotations that contributed most to the heritability of gestational duration were enhancers (Fig. 4) due to the much larger number of enhancer histone marks than promoters in the genome. Our results thus highlight the importance of functional annotations in endometrial stromal cells at GWAS loci for gestational duration.

We next developed a computational procedure, based on fine mapping, to integrate the decidua stromal cell functional maps with a GWAS of gestational duration to identify putative causal variants (Fig. 4A). Because of extensive LD in the human genome, the causal variants driving the associations are unknown at most loci discovered by GWAS. Fine mapping is a Bayesian statistical procedure that takes as input GWAS summary statistics and patterns of LD at trait-associated loci and computes the probability of each variant at a locus to be a causal variant (7). These probabilities, known as posterior inclusion probabilities (PIPs), reflect our confidence of certain SNPs being causal variants. The PIP of a variant ranges from 0 to 1, with 1 indicating full confidence that the SNP is a causal variant. If a region contains a single causal variant, the PIPs of all SNPs in the region should approximately sum to 1.

While fine mapping has been commonly used in identifying putative causal variants from GWAS of complex traits (7), it is often difficult to narrow down causal signals to one or a small number of variants in most GWAS loci. Standard fine mapping treats all SNPs at a locus equally. Recent work suggests that incorporating Bayesian prior probabilities that favor functional SNPs improves fine mapping (8, 42). We posited that integrating functional annotations in pregnancy-relevant cells in a statistical fine-mapping framework would aid in (i) identifying candidate causal variants at each locus associated with gestation duration, (ii) linking those variants to their target genes, and (iii) discovering additional loci and genes associated with gestational duration that may have failed to reach the stringent threshold for significance in GWAS.

We first leveraged the enrichments of DSC annotations to create Bayesian prior probabilities for a variant being causal. On the basis of the results of S-LDSC, we chose H3K27ac, H3K4me1, and pcHi-C interactions from the decidualized cells, and H3K4me3 from untreated cells, as functional genomic annotations to create informative priors using TORUS (42). TORUS takes as input genome-wide summary statistics from GWAS and the functional annotations of SNPs and computes enrichment parameters of annotations, which reflect how much more likely an SNP is a causal variant than randomly chosen SNPs (table S3). SNPs associated with functional annotations are generally assigned higher prior probabilities. In addition, TORUS computes statistical evidence at the level of genomic blocks, defined as the probability that a block (determined by LD) contains at least one causal SNP. Without including any histone marks or chromatin accessibility annotations, TORUS implicated six autosomal blocks in the genome at false discovery rate (FDR) of < 0.05, including five of the six genome-wide significant autosomal loci identified in the GWAS (P < 5 108). One locus on chromosome 3 had an FDR = 0.11 and was therefore not identified by TORUS, and one locus on chromosome 9 that was not identified in the GWAS was implicated by TORUS (data file S3). By including the functional genomic annotations from endometrial stromal cells, the number of high confidence blocks increased to 10, including all 6 that were significant in the gestational duration GWAS and 4 that were not significant in the GWAS (data file S3).

We next performed computational fine mapping on these 10 blocks, with the informative priors learned by TORUS, using sum of single effects (SuSiE) regression (43). Conceptually, SuSiE is a Bayesian version of the stepwise regression analysis commonly used in GWAS (i.e., conditioning on one variant and testing if there is any remaining signal in a region). SuSiE accounts for the uncertainty of causal variants in each step and reports the results in the form of PIPs. Including the priors defined by TORUS using DSC functional annotations significantly improved fine mapping (Fig. 5A, table S3, and data file S4). For example, only one SNP reached PIP > 0.3 across all 10 blocks using the default setting under SuSiE (uniform prior, treating all SNPs in a block equally). This reflects the general uncertainty of pinpointing causal variants due to LD, e.g., a strong GWAS SNP in close LD with nine other SNPs would have PIP about 0.1. By using the annotation-informed priors, eight SNPs in six different blocks reached PIP > 0.3 (Fig. 5A). In some blocks, we were able to fine-map a single high-confidence SNP, e.g., the FOXL2 locus on chromosome 3, while in other blocks, we had considerable uncertainty of the causal variants, as shown by large credible sets, i.e., the minimum set of SNPs to include the causal SNP with 95% probability (Fig. 5B). Table 1 summarizes the most probable causal variants in eight blocks (fine mapping in the remaining two blocks produced large credible sets with no high-PIP SNPs) and their likely target genes based on promoter assignment or chromatin interactions from pcHi-C. We note that our results of the WNT4 locus identified rs3820282 as the likely causal variant. This is consistent with our previous results demonstrating experimentally that the T allele of this SNP disrupts the binding of estrogen receptor 1 (5). This SNP was among the three most likely SNPs in our fine-mapping study, with a PIP of 0.27 (Table 1).

(A) PIPs of SNPs using uniform vs. functional priors in SuSiE (each dot is an SNP). The functional prior of an SNP is based on SNP annotations and is estimated using TORUS. (B) Summary of fine-mapping statistics of all 10 regions. X axis: The size (number of SNPs) of credible set. Y axis: The maximum PIP in a region. We label each region by its top SNP (by PIP) and the likely causal gene, according to Table 1 or the nearest gene of the top SNP. (C) Likely causal variants near HAND2 and their functional annotations. The top panel shows the significance of SNP association in the GWAS and the middle panel shows fine-mapping results (PIPs) in the region. The vertical yellow bar highlights the two SNPs with high PIPs. These SNPs are located in a region annotated with ATAC-seq, H3K27ac, H3K4me1, and H3K4me3 peaks (bottom). This putative enhancer also had increased ATAC-seq, H3K27ac, and H3K4me1 levels in decidualized samples and interacts with the HAND2 promoter (red arc).

Functional annotations are based on data from endometrial stromal cells. We list an annotation if the SNP is located in a sequence with that annotation in either untreated or decidualized condition. Functional prior is the prior probability of an SNP being a causal variant. For an SNP without any functional annotation, its prior probability is 3.6 106. We list the pcHi-C annotation if the SNP is within 1 kb of a region involved in a pcHi-C interaction. We call a gene the target of an SNP if (i) the SNP is located in the promoter (< 1 kb of transcription start site) of that gene or (ii) the promoter of that gene has a pcHi-C interaction with a region within 1 kb of the SNP. In the case of rs147843771 at the FOXL2 locus, the target was defined by literature evidence (69). The number of credible SNPs at each region is shown in Fig. 5B. SNPs in bold are discussed in the text. FOXL2 (69), forkhead box L2; GATA2, GATA-binding protein 2; HAND2, heart and neural crest derivatives expressed 2; KCNAB1, potassium voltage-gated channel subfamily A member regulatory beta subunit 1; WNT4, Wnt family member 4.

We highlight the results from two regions. In both cases, we were able to identify putative risk genes with relatively high confidence, and neither is the nearest gene of lead SNPs in GWAS. In the first case, two adjacent SNPs [311base pair (bp) apart], rs13141656 and rs7663453, on chromosome 4q34 did not reach genome-wide significance in the GWAS (P = 3.9 107 and 4.5 107, respectively). After using functional annotations in decidua-derived stromal cells, the block containing these SNPs was highly significant (TORUS q = 0.02), suggesting the presence of at least one causal variant in this block. The two SNPs together explained most of the PIP signal in the block (PIP 0.38 and 0.33, respectively, Table 1). The two SNPs are located in a region of open chromatin in endometrial stromal cells, with enhancer activity marked by both H3K27ac and H3K4me1 (Fig. 5C). Only 9 of the 129 tissues from the Epigenome Roadmap (11) also had H3K27ac, H3K4me1, or H3K4me3 peaks spanning the rs13141656 locus and only 2 spanning the rs7663453 locus. In addition, this putative enhancer is bound by multiple transcription factors, including GATA2, FOXO1, NR2F2, and PGR, based on ChIP-seq data. The only physical interaction of this enhancer in the pcHi-C data in decidualized stromal cells is with the promoter of the HAND2 gene, located 277 kb away (Fig. 5C). Summing over the PIPs of all SNPs whose nearby sequences interact with HAND2 (heart and neural crest derivatives expressed 2) via chromatin looping gives an even higher probability, 0.89, suggesting that HAND2 is very likely to be the causal gene in this region (table S4). HAND2 is an important transcription factor that mediates the effect of progesterone on uterine epithelium (44). Thus, in this example, we identified a previously unknown locus, the likely causal variant(s), the enhancers they act on, and an outstanding candidate gene for gestational duration and PTB.

The second example focuses on the locus showing a strong GWAS association with gestational duration on chromosome 3q21. The lead SNP, rs144609957 (GWAS P = 4 1013), is located upstream of the EEFSEC (eukaryotic elongation factor, selenocysteine-tRNAspecific) gene. There is considerable uncertainty of the causal variants in this region, with 50 SNPs in the credible set and the lead SNP explaining only a small fraction of signal (PIP = 0.02). Among all 12 SNPs with PIP > 0.01, 11 have functional annotations, most commonly H3K4me1 and pcHi-C interactions. For nine SNPs (first three shown in Table 1), the sequences in which they are located physically interact with the promoter of GATA2 in the pcHi-C data but not with any other promoters in the region (fig. S3). The PIPs of all SNPs in the genomic regions that likely target GATA2 through chromatin looping sum to 0.68 (table S5). Thus, despite uncertainty of causal variants in this region, our results implicate GATA2 as a candidate causal gene in endometrial stromal cells. GATA2 is a master regulator of embryonic development and differentiation of tissue-forming stem cells (45). As support for the possible role of GATA2 in pregnancy, GATA2 deficient mice show defects in embryo implantation and endometrial decidualization (35), making this another excellent candidate causal gene for gestational duration and PTB.

The molecular processes that signal the onset of parturition in human pregnancies, and how perturbation of those processes result in PTB, are largely unknown. Yet, understanding these processes would reveal important insights into the potential causes of adverse pregnancy outcomes, including spontaneous labor before 37 weeks gestation, and potentially lead to the identification of biomarkers and therapeutic targets for PTB. Although it is experimentally challenging to link decidualization processes directly to parturition in humans, it is well accepted that shallow implantation due to suboptimal decidualization is associated with poor pregnancy outcomes in general (4648) and that the decidua is key in triggering parturition (13, 14). Thus far, however, specific genes that perturb decidualization processes and lead to PTB are poorly defined.

Unbiased GWASs do not require prior knowledge of molecular processes underlying disease phenotypes and have the potential to identify novel genes and pathways contributing to common diseases. However, the significant heterogeneity of most common diseases and small effects of most common disease-associated variants lead to the requirement for very large sample sizes (in the tens to hundreds of thousands of cases) to discover more than a handful of associated loci that meet stringent criteria for genome-wide significance. To address this limitation and provide orthogonal evidence for assessment of associations, we characterized the transcriptional and chromatin landscapes in decidua-derived stromal cells and integrated those functional annotations with a GWAS of gestational duration to discover novel loci and genes. The primary motivation for these studies was the notable paucity of genomic and epigenomic functional annotations in pregnancy-relevant primary cells among those studied by large consortia (911). Here, we filled a significant gap by providing maps in untreated and decidualized stromal cells and used these maps for annotating GWAS of pregnancy-related traits.

We chose to focus these studies on endometrial stromal cells because of their central importance in both the establishment and maintenance of pregnancy, as well as their intimate juxtaposition to fetal trophoblast cells throughout pregnancy. Of particular relevance are the roles that decidualized stromal cells play in regulating trophoblast invasion, modulating maternal immune and inflammatory responses at the maternal-fetal interface, and controlling remodeling of the endometrium (48). Defects in all of these processes have been considered a contributing factor to pregnancy disorders (48, 49). Moreover, we showed that the SNPs in regions with endometrial stromal cell functional annotations explained more of the heritability of gestational duration compared to just using baseline annotations. Among all annotations, enhancer marks H3K4me1 (in both decidualized and untreated stromal cells) and H3K27ac (in decidualized cells) were 8- to 10-folds enriched at GWAS loci after adjusting for the general annotations and accounted for 50 to 70% of the GWAS heritability. The lack of complete independence between these marks makes it difficult to delineate their individual effects but, nonetheless, highlights the importance of enhancers and of gene regulation in endometrial stromal cells in modulating the effects of GWAS variants on gestational duration. This is consistent with both the known tissue-specific roles of enhancers and the observation that more than 90% of GWAS loci reside outside of the coding portion of the genome and are enriched in regions of open chromatin and enhancers (12, 41).

Integrating transcriptional and chromatin annotations of gene regulation from MSCs and DSCs improved our ability to discover novel GWAS loci and identify likely causal SNPs and genes associated with gestational duration. We illustrate how our integrated platform identified a novel causal locus and candidate gene (HAND2) associated with gestational duration, as well as refined the annotation of loci that had been previously identified. Our data suggest that in endometrial stromal cells, GATA2 is likely the target gene of enhancers harboring SNPs associated with gestational duration. This does not exclude the possibility that the nearest gene to the associated SNPs, EEFSEC, may be a target gene in other cell types. Both HAND2 (50) and GATA2 (51) are involved in decidualization processes in humans, and perturbations in this process have been linked to poor pregnancy outcomes (4648). Neither GATA2 nor HAND2 was identified as potential candidate genes in previous GWASs of gestational duration, or PTB supports our approach and the importance of using functional annotations from cell types relevant to pregnancy to fine map and identify candidate genes for the pregnancy-related traits. Overall, the integrated analyses performed in this study resulted in the identification of both novel GWAS loci and novel candidate genes for gestational duration, as well as maps of the regulatory architecture of these cells and their response to decidualization.

However, there are some limitations. Our results are based on cells from only three individuals, which may not fully capture the regulatory landscape of endometrial stromal cells. For pcHI-C, we used cells from a single individual to generate the chromatin interactions map. Another limitation is that we focused on only one cell type, albeit one that plays a central role in pregnancy and only one exposure (hormonal induction of decidualization) at one time point (48 hours). Furthermore, it is unclear how our model of in vitro decidualization mimics the endogenous decidualization of endometrial cells during pregnancy. While we chose decidualization as a perturbation to ascertain the dynamic features of functional genomic annotations, we fully anticipate that obtaining annotations in other cell types and in response to other relevant perturbations will improve the ability to identify novel loci, variants, and genes associated with PTB. Future studies that include fetal cells from the placenta and uterine or cervical myometrial cells could reveal additional processes that contribute to gestational duration and PTB, such as those related to fetal signaling and the regulation of labor, respectively. Inclusion of additional exposures, such as trophoblast conditioned media (52) and additional exposure times, may further reveal processes that are pregnancy specific. Second, to maximize power, we focused on a GWAS of gestational duration and not PTB per se. While previous GWAS have shown that all PTB loci were among the gestational age loci (5), we realize that some of the loci that we identified could be related to normal variation in gestational duration and not specifically to PTB. Nonetheless, our findings contribute to our understanding of potential mechanisms underlying the timing of human gestation, about which we still know little. Last, although our ChIP-seq results revealed an association between GATA2 binding and decidualization, confirming the role of this transcription factor in decidual cell biology (53, 54), and studies in murines support its role in endometrial processes (35), we do not yet have direct evidence showing that perturbations in the expression of GATA2, or any of the other target genes identified, influence the timing of parturition in humans. Future studies will be needed to directly implicate the expression of these genes in gestational duration or PTB. Our study highlights the importance of generating functional annotations in pregnancy-relevant cell types to inform GWASs of pregnancy-associated conditions. Our results suggest that the expression of two transcription factors, GATA2 and HAND2, in endometrial stromal cells may regulate transcriptional programs that influence the timing of parturition in humans, which could lead to the identification of biomarkers of or therapeutic targets for PTB.

This study was approved by the Institutional Review Boards at the University of Chicago, Northwestern University, and Duke University Medical School. Informed consent for the use of data collected via questionnaires and clinics was obtained from participants following the recommendations of the ALSPAC (Avon Longitudinal Study of Parents and Children) Ethics and Law Committee at the time. Informed consent for the use of genetic data in the other six GWASs used in this study was also obtained from participants. Details are available in the Supplementary Materials.

Placentas were collected from three African American women (18 years old) who delivered at term (37 weeks) following spontaneous labor; all were vaginal deliveries of singleton pregnancies. Within 1 hour of delivery, 5 cm by 5 cm pieces of the membranes were sampled from a distant location of the rupture site. Pieces were placed in Dulbeccos modified Eagles medium (DMEM)-Hams F12 media containing 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin. Samples were kept at 4C and processed within 24 hours of tissue collection. This study was approved by the Institutional Review Boards at the University of Chicago, Northwestern University, and Duke University Medical School.

Third trimester placental tissue was enzymatically digested by a modification of previously described methods (55, 56). Decidua tissue was gently scraped from chorion, and tissue was enzymatically digested in a solution (1 Hanks balanced salt solution, 20 mM Hepes, 30 mM sodium bicarbonate, and 1% bovine serum albumin fraction V) containing collagenase type IV (200 U/ml; Sigma-Aldrich, C-5138), hyaluronidase type IS (1 mg/ml; Sigma-Aldrich, H-3506), and DNase type IV (0.45 KU/ml, Sigma-Aldrich, D-5025) at 37C, until a single-cell suspension was obtained (usually three rounds of 30 min digestion using fresh digestion media each round). Epithelial cells were removed by filtering through a 75 M nylon membrane and RPMI (Sigma-Aldrich) containing 10% FBS was added for enzyme inactivation. Dissociated cells were collected by centrifugation at 400g for 10 min and washed in RPMI/10% FBS. Erythrocytes were removed by cell pellet incubation with 1 red blood cell lysis buffer (Sigma-Aldrich) for 2.5 min at room temperature. The resulting cells were counted and resuspended in seeding media [1 phenol red-free high-glucose DMEM (Gibco)] supplemented with 10% FBS (Thermo Fisher Scientific), 2 mM l-glutamine (Life Technologies), 1 mM sodium pyruvate (Fisher Scientific), 1 insulin-transferrin-selenium (ITS; Thermo Fisher Scientific), 1% penicillin/streptomycin, and 1 antibiotic-antimycotic (Thermo Fisher Scientific). Dissociated cells were plated into a T75 flask and incubated at 37C and 5% CO2 for 15 to 30 min (enrichment by attachment). The supernatant was carefully removed, and loosely attached cells were discarded. Plates were allowed to grow in fresh media containing 10% charcoal-stripped FBS (CS-FBS), and 1 antibiotic-antimycotic until the plate was 80% confluent. The antibiotic-antimycotic was removed from the culture media after 2 weeks of culture. We obtained >99% vimentin-positive cells after three passages (fig. S4). Cells were expanded, harvested in 0.05% trypsin, and cryopreserved in 10% dimethylsulfoxide culture media for subsequent use. Each cell line was defined as coming from a different sample collection (different pregnancy).

Cells were plated and grown for 2 days in cell culture media (1 phenol red-free high-glucose DMEM, 10% CS-FBS, 2 mM l-glutamine, 1 mM sodium pyruvate, and 1 ITS). After 2 days, cells were treated either with control media (1 phenol red-free high-glucose DMEM, 2% CS-FBS, 2 mM l-glutamine) or decidualization media (1 phenol red-free high-glucose DMEM, 2% CS-FBS, 2 mM % l-glutamine, 0.5 mM 8-Br-cAMP, and 1 M MPA) for 48 hours. Cells were incubated at 37C and 5% CO2 and harvested for ATAC-seq, ChIP-seq, and RNA-seq, and prolactin (PRL) and insulin-like growth factor-binding protein 1 (IGFBP1) mRNA were assessed by quantitative real-time polymerase chain reaction (PCR) before each downstream assay was performed.

Total RNA was extracted from approximately 1 million cells using the AllPrep DNA/RNA Kit (QIAGEN) according to manufacturers instructions. RNA quality (RNA integrity number) and concentration was assessed by Bioanalyzer 2100 (Agilent technology). RNA-seq libraries were generated by a TruSeq stranded total RNA library prep kit (Illumina) and TruSeq RNA CD Index Plate.

For ChIP experiments, cells were cross-linked by adding to the media 37% formaldehyde to a final concentration of 1%, gently mixed, incubated for 10 min, and quenched for 5 min with 2.5 M glycine for a final of 0.125 M per plate. Cells were washed using cold 1 phosphate-buffered saline and scraped in 15 ml of cold Farnham lysis buffer and protease inhibitor (Roche, 11836145001), and cell pellets were flash frozen and kept at 80C. Thawed pellets were resuspended in radioimmunoprecipitation assay buffer on ice, aliquoted into 20 million cells per tube, and sonicated by Bioruptor (three 15-min rounds of 30 s ON, 30 s). ChIP was performed on 10 million cells using antibodies to H3K27ac, H3K4me3, and H3K4me1 histone marks (ab4729/lot no. GR274237, ab8580/lot no. GR273043, and ab8895/lot no. GR262515, respectively). M-280 sheep anti-rabbit immunoglobulin G Dynabeads (Invitrogen, 11203D) was used for chromatin immunoprecipitation. DNA was purified using the Qiagen MinElute PCR Purification Kit, quantified by Qubit, and prepared for sequencing using the Kapa Hyper Prep Kit. All libraries were pooled to 10 nM per sample before sequencing.

Approximately, 50,000 cells were harvested and used for ATAC-seq library preparation as described in the Fast-ATAC protocol (57). ATAC-seq libraries were uniquely indexed with Nextera PCR Primers and amplified with 9 to 12 cycles of PCR amplification. Amplified DNA fragments were purified with 0.8:1 ratio of Agencourt AMPure XP (Beckman Coulter) to sample. Libraries were quantified by Qubit, and size distribution was inspected by Bioanalyzer (Agilent Genomic DNA chip, Agilent Technologies). All libraries were pooled to 10 nM per sample before sequencing.

In situ Hi-C was performed as described previously (58). Briefly, 5 million decidualized cells were treated with formaldehyde 1% to cross-link interacting DNA loci. Cross-linked chromatin was treated with lysed and digested with MboI endonuclease (New England Biolabs). Subsequently, the restriction fragment overhangs were filled in and the DNA ends were marked with biotin-14-dATP (Life Technologies). The biotin-labeled DNA was sheared and pulled down using Dynabeads MyOne Stretavidin T1 beads (Life Technologies, 65602) and prepared for Illumina paired-end sequencing. The in situ Hi-C library was amplified directly off of the T1 beads with nine cycles of PCR using Illumina primers and protocol (Illumina, 2007). Promoter capture was performed as described previously (39). The Hi-C library was hybridized to 81,735 biotinylated 120-bp custom RNA oligomers (Custom Array) targeting promoter regions (four probes/RefSeq transcription start sites). After hybridization, postcapture PCR was performed on the DNA bound to the beads via biotinylated RNA.

Read counts per gene were calculated with Salmon (59) version 0.12.0 on transcripts from human Gencode release 19 (ftp://ftp.ebi.ac.uk/pub/databases/gencode/Gencode_human/release_19/gencode.v19.pc_transcripts.fa.gz and ftp://ftp.ebi.ac.uk/pub/databases/gencode/Gencode_human/release_19/gencode.v19.lncRNA_transcripts.fa.gz). Estimated counts were used in exploratory analysis (transformed with DESeq2s rlog function) and in DESeq2 (25) version 1.24.0 to identify differentially expressed genes (adjusted P 0.05 and absolute fold change of 1.2). After observing that replicates for each cell lines clustered together, we pooled reads for each cell line, combining three decidualization experiments in each sample. We then performed a paired analysis to obtain genes that were differentially expressed between untreated and decidualized samples. The six samples clustered by treatment and by cell line and analysis with svaseq (60) showed that the two surrogate variables identified correlated with cell line, and therefore, a paired analysis was enough to correct the data.

Similarly to RNA-seq, we pooled reads from replicates for each cell line. We called peaks for each of the six samples using MACS2 and converted peak coordinates into 100-bp contiguous bins. Bins covered by less than 60% of their extension were excluded. To identify reproducible peaks, we only kept bins that were present in at least two of the three cell lines in each condition, allowing for condition-specific peaks. See table S7 for an assessment of the contribution of each cell line to the universe of peaks obtained. We then merged all adjacent bins, expanding them back into longer peaks. We counted the number of reads in all peaks and in all samples and compared the read counts using DESeq2 (adjusted P < 0.05 and absolute fold change >1.2).

The P values in Fig. 2B were calculated with a chi-square test of the number of peaks with increased or decreased numbers of reads observed and an expected probability based on the number of peaks in each category for each dataset. Bonferroni correction was performed to correct for multiple testing.

ChIP-seq reads were downloaded from National Center for Biotechnology Information Gene Expression Omnibus and processed locally. HOMER 4.9 (63) was used to call peaks for the following samples: PGR (GSE94038); NR2F2 (GSE52008); FOSL2 (GSE94038), FOXO1 (GSE94037); and NR2F2 input (GSE52008); and FOXO1, PGR, and FOSL2 input (GSE94038).

Reproducible peaks were converted into 100 bp bins and those with >60% of their extension covered by a peak was retained. Common bins were counted, and the number of counts was plotted with UpSetR 1.4.0.

We used HOMER 4.9 to identify DNA binding motifs enriched in peaks with parameters -len 8,10,12 -size 200 -mask.

Enrichment was calculated as the observed number of overlapping peaks divided by the expected number of overlapping peaks using bedtools intersectBed with a 1 bp minimum. The expected number of overlapping peaks was obtained by averaging 100 random samples of peaks with bedtools shuffle excluding gaps annotated by the University of California, Santa Cruz Genome Browser (64). While shuffling peaks does not account for mapping and other biases that make peak locations nonuniform and may result in overestimation of enrichment, our results are limited to comparisons between enrichments, which should cancel any biases.

We used HiCUP v0.5.9 (65) to align and filter Hi-C reads. HiCUP used bowtie2 version 2.2.3 to align reads. Unique reads were used as input by CHiCAGO (66) version 1.2.0, and significant interactions were called with default parameters. We only kept interactions identified by CHiCAGO that were in cis and with an end located at least 10 kb from a capture probe.

To pair peaks using pcHi-C, significant interactions identified by CHiCAGO that overlapped an ATAC-seq or ChIP-seq peak and were less than 300 kb away from a promoter were used. We chose 300 kb because the mean distance between interacting promoters and other regions was 280 kb (median, 200 kb). To pair peaks to the nearest gene, BEDTools closest -t first -d was used to find the gene closest to a peak, up to 300 kb away. To pair peaks to a random gene, all genes up to 300 kb from a peak were selected and one gene was randomly assigned to each peak. For each of these sets of pairs, we calculated the fraction of peak/gene pairs that had the same direction of change according to differential read count analysis with DESeq2, of the total number of peak/gene pairs. Only genes expressed at >1 transcript per million across all samples were used in the nearest and random gene assignments.

P values were calculated with a chi-square test comparing the number of cases in the matched and unmatched categories observed in the random set (average from 200 iterations) and in the two peak/gene pairing methods: nearest gene and pcHi-C interactions.

The GWAS results used in this study was an extension of our previously published results (5). Like our previous study, we used summary results from 23andMe, which were obtained from GWAS of gestational duration in 42,121 mothers of European ancestry. In addition, we performed GWA analyses in 14,263 European mothers from six academic datasets. To increase the power of GWA discovery, we performed meta-analysis between the results from 23andMe and the results from the six datasets. See the Supplementary Materials for a full description of the GWAS.

We assessed how much of the heritability of gestational duration is contained within ATAC-seq, H3K4me1, H3K4me3, H3K27ac, and pcHi-C peaks using S-LDSC (41). S-LDSC is a generalization of LD score regression, a method for estimating the heritability of a trait using SNP-level GWAS summary statistics and SNP-level estimates of the amount of genetic variation tagged at each variant, known as LD scores. Under the LD score regression model, the expected value of the GWAS summary statistic for a variant (specifically, the expected value of the 2 statistic) is a linear function of the LD score at that site, and h2, the per-SNP heritability, and a an intercept parameter. Under the S-LDSC model, rather than estimating a single per-SNP heritability parameter, a parameter is estimated for each of several functional annotations. In a standard S-LDSC analysis, user-provided annotations are combined with a baseline set of genomic annotations from publicly available datasets. For this analysis, LD scores were calculated using the peaks identified as reproducible across either treated or untreated samples as annotations and the genotype data from the European individuals from phase 3 of the 1000 Genomes project (obtained from the Price Lab website: https://alkesgroup.broadinstitute.org/LDSCORE/) as a reference LD panel, using only the HapMap3 SNP list (also from the Price lab website). S-LDSC was performed on the gestational duration GWAS using the endometrial-tissue derived LD scores and the baseline LD scores contained in version 2.2 of the LD score regression baseline LD model. We include all annotations from the baseline LD model except those flanking annotations. This resulted in a total of 64 baseline annotations used in our S-LDSC analysis.

Fine mapping proceeded in three stages. In the first stage, we partitioned the genome into 1703 regions approximately independent regions using breakpoints derived by Berisa et al. (40). Next, we constructed an SNP-level prior probability of being causal variant, informed by the functional genomic data that we collected. We used a Bayesian hierarchical model [TORUS (42)]. TORUS takes as input GWAS summary statistics and genomic annotations and estimates the extent to which SNPs with functional genomic annotations are likely to be causal for a trait of interest. Specifically, under TORUS, each SNP has a small prior probability of being a causal variant, which is a logistic function of the annotations of the SNP. Then, TORUS estimates the parameters of this logistic function using genome-wide summary statistics. Once these parameters are estimated, each SNP will have a prior causal probability based on its unique functional annotations. We ran TORUS with the gestational age GWAS summary statistics and the reproducible H3K27ac and H3K4me1 peaks from the treated samples along with the pcHi-C contact regions to obtain an SNP-level prior.

Last, fine mapping was performed using a summary statistics-based version of the sum of single effects model (43) using 1000 Genome as reference panel. SuSiE (as implemented in the R package susieR) was run on the 10 regions believed to have one or more causal variants with an FDR of 0.1 as estimated by TORUS. For each region, SuSiE was run with a uniform prior (default setting of SuSiE) and with an informed prior learned by TORUS. The parameter L of SuSiE (maximum number of causal variants) is set at 1 when running SuSiE (67, 68).

H3K27ac, H3K4me1, and H4K4me3 histone modification peak coordinates were downloaded from the Epigenome Roadmap data website, and bedtools intersect was used to find peaks that overlapped SNPs coordinates.

Read the rest here:
Transcriptome and regulatory maps of decidua-derived stromal cells inform gene discovery in preterm birth - Science Advances

Read More...

Therapy Options Compared for the Treatment of Recurrent DLBCL – Targeted Oncology

Friday, December 4th, 2020

Loretta J. Nastoupil, MD, associate professor, director, Lymphoma, Outcomes Database Section chief, New Drug Development Department of Lymphoma/Myeloma ,Division of Cancer Medicine at The University of Texas MD Anderson Cancer Center, discussed the use of chimeric antigen receptor (CAR) T-cell therapy in patients with diffuse large B-cell lymphoma.

The discussion occurred among a group of oncologist during a Targeted Oncology Case Base Peer Perspective event.

Targeted Oncology: What clinical trial data support the use of CAR T-cell therapy for this patient?

NASTOUPIL: The ZUMA-1 study [NCT02348216] enrolled patients with refractory large cell lymphoma and included about 30% of patients with double-hit features. [It] also included [patients with] primary mediastinal transformed follicular lymphoma. There was a relatively small sample size of 101 patients. This was a single-arm, phase 2 [design]. However, they had a high success rate in terms of enrollment and treatment of patients where 91% of the patients who were enrolled received cell therapy.

ZUMA-1 [examined axicabtagene ciloleucel (axi-cel; Yescarta)], an autologous CD19-directed CAR T-cell therapy. It has a CD28 costimulatory molecule. The lymphocyte depleting regimen is high in this study: 500 mg/m2 of cyclophosphamide and 30 mg/m2 of fludarabine for 3 consecutive days.

The median progress-free survival [PFS] in this study was about 6 months [5.9 months; 95% CI, 3.3-15.0]. The best objective response rate [ORR] was 83% and best complete response [CR] rate was 58%.

There was a stark drop-off and then a flattening of the [Kaplan-Meier PFS] curve. [Experience has shown that] theres about 40%, to maybe as high as 50%, of patients who can be cured with this, but its hard to identify who they are. The patients who dont respond generally do poorly within the first 3 to 6 months.

The JULIET study [NCT02445248] was a bit different. Its a larger multicenter study [that examined tisagenlecleucel (Kymriah)]. It was a much longer time from enrollment to infusion of CAR T cells; as a result, bridging therapy was allowed. They also included relapsed, not just refractory, patients, and they excluded patients with primary mediastinal disease.2

There [were] lower response rates [in] this study population of 52%. The CR rate, though, is notable at 40%. The patients who had CR had durability of that CR. About 40% of the patients had a meaningful outcome with this CAR-T cell therapy, which was a 4-1BB construct. This is an autologous [product] with a CD19 target.

The efficacy is hard to compare across the studies because they were different in terms of patient eligibility and trial conduct. However, my general sense is that the vast majority of patients have about a 40%, to maybe as high as 50%, meaningful CR that is durable, and the overall survival [OS] is quite notable. With ZUMA-1, the median OS settles in at 27.1 months.

Are there any CAR T-cell therapies that may be added to the list of available products for these patients?

Liso-cel [lisocabtagene maraleucel] is the third [CAR T-cell product that is] anticipated for FDA approval. Thats also a 41BB construct similar to tisagenlecleucel. Its different in that theres a fixed CD4 to CD8 ratio and a longer time for manufacturing in comparison to axi-cel. But again, there is meaningful efficacy.3

How does the toxicity compare between these agents?

The toxicity looks to be different, but different grading systems were applied in the studies. When you apply the same rating system retrospectively, patients have fewer grade 3 or higher cytokine release syndrome in the JULIET study than what was initially reported.4

Axi-cel tends to have the highest rate of grade 3 or higher neurotoxicity and liso-cel tends to have the most favorable toxicity profile. Though because its the third [agent to become available], weve [become] much better at identifying and mitigating some of these acute toxicities.

There are differences, in my opinion, across the 3 constructs in terms of safety, despite the efficacy being quite similar.

Why are these data important to review?

We know CAR-T cell therapy has transformed outcomes for about 40% of patients; however, its logistically challenging....Weve struggled with identifying what makes a good CAR-T cell candidate outside of having chemotherapy-refractory disease and being in a third-line setting.

What other therapy options could be used in this patient if she is not a candidate for CAR T-cell therapy?

Polatuzumab vedotin [Polivy] in combination with bendamustine and rituximab [BR] was approved based on a randomized phase 2 study [NCT02257567].5 Of note, this was a study that included both follicular and large cell lymphoma. However, the follicular [cohort showed] a negative result. There was no significant impact with the addition of polatuzumab vedotin to BR, which is not necessarily surprising given that bendamustine is a very active agent for follicular lymphoma.

It is not an active agent in large cell lymphoma. One of the reasons why it was included in this study design is because the potential for challenge with polatuzumab was identifying an agent in third-line large cell lymphoma setting where you wouldnt have additive.

Please describe the trial that led to the approval of polatuzumab vedotin.

For baseline characteristics, and this is specifically looking at the patients with large cell lymphoma, the median age was 67 years [range, 33-86] in the polatuzumab/BR arm versus 71 years [range, 30-84] in the BR-only arm. In terms of median number of prior lines of therapy, it was 2 for both arms. In terms of prior stem cell transplant, about 25% had undergone transplant in the polatuzumab-BR arm versus 15% in the BR-arm.

ORR by independent review with the polatuzumab-BR was much higher versus the BR-only arm [45% versus 17.5%].6

Importantly, the median PFS was significantly different. If patients received polatuzumab, the median PFS was 9.5 months versus 3.7 months in the control arm [0.36; 95% CI, 0.21-0.63; P < .001].

The important message is that there werent any subgroups that benefited from BR alone, and this included criteria such as bulky disease, number of prior lines of therapy, and duration of response with the prior line of therapy.

The OS was significantly better if you had polatuzumab plus BR, with a median of 12.4 months versus 4.7 months [HR, 0.42; 95% CI, 0.24-0.75; P = .002]. Similarly, there were no subgroups that tended to benefit from receiving BR alone.

What was the toxicity profile of this regimen for patients?

[It is] important to note that peripheral neuropathy, which is an adverse event associated with this antibody-drug conjugant, was mostly reported as grade 1 events. [The effect was] slightly additive if you had the BR and polatuzumab versus no polatuzumab.

In terms of neutropenia, grade 3/4 events with the combination occurred in 46.2% of patients, and thats higher when compared with BR alone [33.3%]. However, febrile neutropenia events were no different between arms [10.3% vs 12.8%].

References:

1. Neelapu SS, Locke FL, Bartlett NL, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017;377(26):2531-2544. doi:10.1056/NEJMoa1707447

2. Schuster SJ, Bishop MR, Tam CS, et al; JULIET Investigators. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med. 2019;380(1):45-56. doi:10.1056/NEJMoa1804980

3. Abramson JS, Palomba ML, Gordon LI, et al. Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study. Lancet. 2020;396(10254):839-852. doi:10.1016/S0140-6736(20)31366-0

4. Abramson JS. Anti-CD19 CAR T-cell therapy for B-cell non-Hodgkin lymphoma. Transfus Med Rev. 2020;34(1):29-33. doi:10.1016/j.tmrv.2019.08.003

5. FDA approves polatuzumab vedotin-piiq for diffuse large B-cell lymphoma. FDA. June 10, 2019. Accessed November 13, 2020. https://bit.ly/2IBZGrv

6. Sehn LH, Herrera AF, Flowers CR, et al. Polatuzumab vedotin in relapsed or refractory diffuse large B-cell lymphoma. J Clin Oncol. 2020;38(2):155-165. doi:10.1200/JCO.19.00172

Link:
Therapy Options Compared for the Treatment of Recurrent DLBCL - Targeted Oncology

Read More...

Retracing the evolutionary emergence of thymopoiesis – Science Advances

Friday, December 4th, 2020

INTRODUCTION

The adaptive immune system arguably represents one of the major evolutionary novelties that distinguish vertebrates from their nonvertebrate ancestors. About half a billion years ago, specialized cell types, such as lymphocytes, and new organs, such as the thymus, emerged, contributing to the radical redesign of animal immunity (13). These morphological and cytological innovations likely occurred in the ancestor common to all vertebrates, since they are present in both of the two extant groups of vertebrates, the jawless and jawed vertebrates (3). Additional novelties, such as the chemokine and chemokine receptor systems, enabled new types of cellular interactions, for instance, between hematopoietic progenitor cells and the emerging thymopoietic tissue (4). The thymic microenvironment provides distinct molecular cues such as Notch ligands, chemokines, and cytokines that interact in a synergistic, context-dependent, and hierarchical manner and, in this way, determine the outcome of hematopoietic precursor cell differentiation (5). The key importance of the thymic microenvironment for T cell development was first revealed through studies of rodents homozygous for mutations at the nude locus (68), which disrupt thymus development, causing animals to be immune deficient. The nude locus encodes a transcription factor of the forkhead class (9), now designated as Foxn1. Subsequent studies in mice indicated that the Foxn1 gene is dispensable for the formation of the thymic anlage during embryonic development (8) but is required for the subsequent steps of differentiation of primitive precursor cells into the typical cortical and medullary subsets of the thymic epithelium (10). Foxn1-expressing thymic epithelial precursors (11) are bipotent (12, 13), with each cell able to give rise to a self-organizing thymopoietic unit containing cortical and medullary compartments capable of supporting T cell development (12). Thus, in the absence of Foxn1, the master regulator of the thymic epithelium in mammals, thymus differentiation is aborted, and T cell development fails (9). Attempts to identify the key functional elements of the thymic epithelium through transgenic expression of candidate genes has highlighted the roles of the chemokines Cxcl12 and Ccl25, the cytokine/stem cell factor Scf, and the Notch ligand Dll4 (5), which direct the early steps of T cell development in the thymic microenvironment. However, the individual components of the genetic network downstream of Foxn1 that regulates the emergence of distinct subsets of thymic epithelial cells (TECs) are unknown.

So far, it has not been possible to reconstruct the nature and evolutionary sequence of the steps that gave rise to a functional thymus, as no extant species representing intermediate phylogenetic stages along the transition from innate to adaptive immunity are available for analysis. To begin to address this problem, we have developed an in vivo reconstruction strategy that rests on the exchange of the mammalian version of the Foxn1 transcription factor with its evolutionarily distant relatives of nonvertebrate and vertebrate origin. Since the decisions controlling alternative differentiation pathways in the hematopoietic system are strongly influenced by extrinsic cues (5, 1417), we hypothesized that it should be possible to directly examine and compare the thymopoietic properties of distinct types of epithelial microenvironments in the thymus as a result of the activities of different Foxn1/4 family members.

Foxn1 and its paralog Foxn4 comprise a distinct two-member family of vertebrate wing-helix transcription factors that recognize a unique (G+C)-rich DNA target sequence, distinguished by the core tetranucleotide sequence, 5-ACGC (18, 19). This sequence is also recognized by the DNA binding domain (fig. S1A) encoded by the Foxn4 gene of amphioxus (19), indicating that the evolutionary conservation of the protein sequence of the DNA binding domains encoded by this gene family (4) is mirrored in identical target recognition sequences. In addition to the centrally located DNA binding domain, Foxn1/4 transcription factors exhibit N-terminal domains of variable lengths and acidic transcriptional activation domains (20) in their C-terminal region (fig. S1A); of note, the activation domains are functionally interchangeable, as indicated by the fact that the C terminus of the amphioxus (lancelet) Foxn4 protein can replace that of the mouse Foxn1 protein in in vitro assays of transcriptional activation (21). These observations suggest that the N-terminal regions of Foxn1/4 proteins have been important targets for evolutionary modifications, possibly related to changes in function.

To determine the thymopoietic properties of different members of the Foxn1/4 transcription factor family in the context of the mouse hematopoietic system, we have developed a generic transgenic replacement strategy. It begins with Foxn1-deficient mice, in which the thymic epithelium is functionally inactive (10); it is important to note that, because immature progenitor cells persist in the organ anlage, a functional thymus can be formed upon reactivation of the Foxn1 function (12). We then introduce a transgenic construct that contains all regulatory sequences of the mouse Foxn1 gene (fig. S1B) that are necessary to direct tissue-specific and orthotopic expression of any complementary DNA (cDNA) to the endogenous Foxn1 expression domains into this Foxn1-deficient background. A previous study using this system demonstrated that the expression of the cognate mouse Foxn1 cDNA rescued the pleiotropic nude phenotype in the thymus and the skin (22), thus functionally validating the replacement strategy in vivo. In the first application of this method, we showed that the mouse Foxn4 transcription factor gene [which is paralogous to mouse Foxn1 (4), although not expressed in the mouse thymic epithelium (22)] is nonetheless capable of supporting a degree of lymphopoiesis in the reconstructed thymi (22). These results encouraged us to extend our studies to Foxn1/4 family members (fig. S1C) identifiable in extant representatives of evolutionarily more ancient chordates, using the phenotypes of mouse Foxn1 and mouse Foxn4 replacements as references.

For centuries, biologists have debated the evolutionary origin of vertebrates (23); the current scenario of chordate taxa suggests that lancelets are the sister group to tunicates and vertebrates (24, 25). This phylogenetic relationship is mirrored in our analysis of the chordate Foxn1/4 gene family, which suggests that an ancestral metazoan Foxn4 gene gave rise to the Foxn1 and Foxn4 genes of vertebrates (Fig. 1 and fig. S2) (4). Although tunicates are considered to be phylogenetically closest to vertebrates (24, 25), we chose the Foxn4 gene of amphioxus for our functional analyses, because tunicate development is substantially secondarily modified (26). Hence, to understand the role of Foxn4 in the emergence of specific aspects of the vertebrate body plan, lancelets appeared to be better proxies than tunicates. Moreover, we had previously shown that the amphioxus Foxn4 gene is expressed in the pharyngeal endoderm, the future site of the thymic epithelium in vertebrates (4). For the present experiments, we focused on one species of lancelets (Branchiostoma lanceolatum). To study vertebrate-specific aspects of the Foxn1/4 gene family, we turned to an extant representative of the most ancient group of jawed vertebrates, cartilaginous fishes. To this end, we identified and isolated the Foxn1 and Foxn4 genes of elephant shark (Callorhinchus milii) for our replacement studies.

Vertebrate Foxn1 and Foxn4 clades recapitulate the known phylogenetic relationships of vertebrates (representatives of mammals, monotremes/marsupials, birds, reptiles, amphibians, and bony and cartilaginous fishes are depicted). The Foxn1 proteins in vertebrates form a monophyletic clade. The Foxn4 proteins are paraphyletic in vertebrates and in tunicates. The Foxn4 proteins in lancelets form the base of the tree. The support from 1000 bootstrap replicates is shown as color-coded branches. The vertebrate Foxn1 and Foxn4 clades are boxed in different colors. The scale is in units of average amino acid substitutions per site.

To reconstruct the functional changes that occurred along the evolutionary trajectory of the thymic microenvironment in vertebrates, we examined the developmental fate of mouse hematopoietic progenitors in the different types of thymic microenvironments. Since the Foxn1-deficient thymic epithelium fails to differentiate and does not support lymphoid development (fig. S3) (69), any lymphopoietic activity in the reconstituted thymic microenvironment must be driven by the expression of the respective Foxn1/4 family member under study. The transgenic thymi were examined both for their lymphopoietic capacity and the characteristics of the epithelial microenvironment. Whereas the hematopoietic compartment was analyzed by cell surface markers and in situ analysis of tissue sections, TECs were additionally characterized by RNA sequencing (RNA-seq). Guided by our previous work (22), we focused our attention on three major phenotypic aspects of the thymi that were reconstituted by the Foxn1/4 gene family members of amphioxus and shark. With regards to the composition of the thymic microenvironment, we specifically addressed the question of whether they are capable of supporting the formation of a distinct medullary area, a key compartment associated with the selection of a self-tolerant T cell repertoire (27). With respect to the lymphopoietic properties of the reconstructed thymi, we paid particular attention to their capacity to support the differentiation of thymocytes throughout the known developmental trajectory (17); moreover, we scored the presence and localization of immature and mature B cells, since our previous studies indicated a propensity of the mouse Foxn4 gene to support B cell poiesis when expressed in the thymic epithelium (22), a capacity that the wild-type mouse thymus lacks.

We first examined the thymopoietic capacity of lancelet Foxn4 (Bl_Foxn4), the sole family member of the Foxn1/4 gene family in the genome of the cephalochordate B. lanceolatum. Lancelets lack an adaptive immune system, although cytological evidence for the presence of lymphocyte-like cells has been reported in some species (28, 29). Bl_Foxn4 exhibits some thymopoietic activity. After replacement of mouse Foxn1 (Mus musculus, hereafter Mm_Foxn1) with Bl_Foxn4, the transgenic thymic microenvironment predominantly harbors CD4+CD8+ double-positive (DP) immature thymocytes (Fig. 2A), although their number amounts to only about 1% of that in wild-type thymi (Fig. 2B). Two other features distinguish the hematopoietic compartment of Bl_Foxn4 thymi from the corresponding wild-type situation. First, very few, if any, single-positive T cells are detectable (Fig. 2A), and second, the CD4CD8 double-negative compartment is much larger than in wild-type mice (Fig. 2A). Notably, the absolute number of CD45+CD4CD8CD19+B220+IgM+CD93 mature B cells present in Bl_Foxn4 thymi (Fig. 2C) is only 10-fold lower than that in wild-type thymi (Fig. 2D), indicating that, relative to a mouse wild-type thymus, the transgenic microenvironment tends to be more favorable for mature B cells than for immature T cells.

(A) Flow cytometric analysis of CD45+ thymocytes, stained for T cell markers; wt (n = 6) and Bl_Foxn4 (n = 7). (B) Absolute numbers of total thymocytes and CD4+CD8+ DP T cells in wt (n = 7) and Bl_Foxn4 transgenic thymi (n = 6); ***P < 0.001; two-tailed t test for both groups. (C) Flow cytometric analysis of CD45+ thymocytes and B cell markers; wt (n = 6) and Bl_Foxn4 (n = 7). (D) Absolute numbers of immature CD45+CD4CD8CD19+IgMCD93+ B cells; P = 0.1058, two-tailed t test; wt (n = 6) and Bl_Foxn4 (n = 7). (E) Ly51 expression and UEA1 binding on EpCAM+CD45 TECs; wt (n = 5) and Bl_Foxn4 (n = 5). (F) Heatmap of differential gene expression patterns of TECs. Genes whose expression is associated with particular TEC subsets are indicated (green, cTEC-like; red, mTEC-like; blue, mature cTEC); analysis based on 18,808 protein-coding genes. Scale refers to the percentage of maximum and minimum values of transcript counts of individual genes. (G) Total numbers of CD45EpCAM+ TECs; wt (n = 5) and Bl_Foxn4 (n = 8). (H and I) T and B cell poietic indices calculated from ratios of means (SDs correspond to propagated errors); ***P < 0.001 two tailed t test.

The unique lymphoid signature in the Bl_Foxn4-reconstituted thymi is the result of a microenvironment characterized by epithelial cysts (fig. S4, A and B), dominated by cells reminiscent of cortical TECs (cTECs), as determined by cell surface phenotype (EpCAM+Ly51+UEA1) (Fig. 2E) and the keratin signature (K8+K5) in tissue sections (fig. S4A); we note a conspicuous lack of medullary TECs (mTECs) both in flow cytometric (EpCAM+Ly51UEA1+) (Fig. 2E) and immunohistological (K8K5+) (fig. S4A) analyses. Accordingly, the epithelium has a global transcriptional profile heavily skewed toward a cTEC signature (high levels of Bmp4, Psmb11, Cxcl12, and Dll4 genes), with low levels of genes typically associated with mTECs (Trpm5 and Aire) (30); of note, the near-complete absence of mature TECs is also reflected in low levels of Mhc2 gene expression (Fig. 2F). In the wild-type thymus, the Aire gene is expressed in the CD80+MHCIIhi subset of mature mTEC and involved in the regulation of promiscuous gene expression that is required for proper negative selection of autoreactive T cells (31).

Overall, this constellation gives rise to a low thymopoietic index (calculated as the ratio of hematopoietic cells to TECs) for immature CD4+CD8+ T cells; the capacity of Bl_Foxn4 thymi to generate these immature T cells is two orders of magnitude lower than that of wild-type thymi, compatible with the immature status of the transgenic TECs. Because immature CD45+CD4CD8CD19+B220+IgMCD93+ (henceforth IgMCD93+) B cells are barely detectable in the transgenic thymi (Fig. 2, G to I), the reduction of B poietic capacity can be less confidently determined but appears to be at least 10-fold. Collectively, mice expressing Bl_Foxn4 instead of Mm_Foxn1 in TECs fail to support proper T cell development in the thymus, lack appreciable B cellgenerative capacity, and are most likely incapable of executing the necessary positive and negative selection steps that normally give rise to a self-tolerant repertoire of mature T cells. As a consequence, transgenic mice rapidly (fig. S4C) succumb to an inflammatory syndrome, chiefly affecting the intestine (fig. S4D), occasionally accompanied by vitiligo (fig. S4E). Nonetheless, our data indicate that Bl_Foxn4 is preadapted to support the early stages of T lymphopoiesis when expressed in the context of a vertebrate immune system, attesting to the strong evolutionary conservation of the Notch signaling pathway in regulating the differentiation of hematopoietic precursors [reviewed in (14)].

To assess the functional changes associated with the emergence of a vertebrate form of the Foxn4 gene, we next examined the lymphopoietic capacity of the Foxn4 gene of an extant representative of an evolutionarily ancient group of jawed vertebrates (Fig. 1 and figs. S1 and S2). When the thymic epithelium expresses the Foxn4 gene of the cartilaginous fish C. milii (Cm_Foxn4) instead of Mm_Foxn1, the lymphoid compartment (Fig. 3, A to E) features a large fraction of IgMCD93+ immature B cells, which amounts to about 10% of all hematopoietic cells. This represents an increase of three orders of magnitude when compared to the fraction of immature B cells among the thymocytes of wild-type mice, which averages about 0.01% of all thymocytes. In absolute terms, the Cm_Foxn4 thymi harbor only about 1% of the hematopoietic cell numbers in wild-type mice. Despite the comparatively low number of T cells in the transgenic thymi, their differentiation appears to proceed normally, as reflected by the presence of DP and single-positive thymocytes and the absence of systemic inflammation. These results suggest that the Cm_Foxn4 thymic microenvironment is conducive to both T and B cell differentiation (Fig. 3, F to H); this type of unusual lymphoid bipotency was previously observed in Mm_Foxn4 transgenic thymi (22).

(A to C) Total numbers of CD45+ hematopoietic cells {wt (n = 20), Cm_Foxn4 (n = 7), Cm_Foxn1 (n = 11), and Cm_Foxn1/Cm_Foxn4 double transgenics [Cm_dtg (n = 5)]}, DP T cells [wt (n = 20), Cm_Foxn4 (n = 7), Cm_Foxn1 (n = 11), and Cm_dtg (n = 5)], and CD93+ immature B cells [wt (n = 13), Cm_Foxn4 (n = 7), Cm_Foxn1 (n = 7), and Cm_dtg (n = 5)] in thymi of mice with the indicated genotypes. (D and E) Representative flow cytometric profiles for mice summarized in (A to C). (D) wt (n = 16), Cm_Foxn4 (n = 7), Cm_Foxn1 (n = 9), Cm_dtg (n = 5). (E) wt (n = 13), Cm_Foxn4 (n = 7), Cm_Foxn1 (n = 7), and Cm_dtg (n = 5). (F) Total numbers of CD45EpCAM+ TECs; wt (n = 17), Cm_Foxn4 (n = 6), Cm_Foxn1 (n = 11), and Cm_dtg (n = 5). (G) T cell poietic indices; wt (n = 20), Cm_Foxn4 (n = 6), Cm_Foxn1 (n = 11), and Cm_dtg (n = 5). (H) B cell poietic indices; wt (n = 13), Cm_Foxn4 (n = 6), Cm_Foxn1 (n = 7), and Cm_dtg (n = 5). In (A) to (C) and (F) to (H), each data point represents one mouse. ***P < 0.001; one-way ANOVA with Tukeys multiple comparison test in (A) to (C) and (F) to (H).

Compared to the situation in Bl_Foxn4 transgenic mice, the epithelial microenvironment of Cm_Foxn4 thymi exhibits the cell surface phenotype (Fig. 4A) and the keratin (Fig. 4B) and gene (Fig. 4C) expression patterns of a more mature cTEC compartment; this is particularly evident from the much higher expression levels of Prss16 and Ccl25 genes that are elevated to wild-type levels (Fig. 4C).

(A) Representative flow cytometric analyses of Ly51 expression and UEA1 binding on TECs for mice; wt (n = 22), Cm_Foxn4 (n = 6), Cm_Foxn1 (n = 11), and Cm_dtg (n = 5). (B) Epithelial microenvironment of reconstructed thymi resolved by keratin 5 (K5) (in green) and K8 (in red) staining; 4-week-old mice. m, medulla; c, cortex. Note the small size of the Cm_Foxn4-reconstructed thymus. (C) Differential gene expression patterns in TEC transcriptomes relative to wild-type mice; Bl_Foxn4 (n = 4), Cm_Foxn4 (n = 4), and wt (n = 3). (D) Localization of Aire+ cells relative to cortical (K8) and medullary (K5) areas. (E) Differential gene expression patterns in TEC transcriptomes of transgenic relative to wild-type mice; wt (n = 3), Mm_Foxn4 (n = 6), and Cm_Foxn4 (n = 4). (F) Localization of B220+ B cells (in green) adjacent to ER-TR7+ mesenchyme (in red). PVS, perivascular space. (G) Differential gene expression patterns in TEC transcriptomes of transgenic relative to wild-type mice; wt (n = 3), Cm_Foxn4 (n = 4), Cm_Foxn1 (n = 3), and Cm_dtg (n = 5). In (C), (E), and (G), each data point represents the average value of at least three mice; all values are significantly different from the wild-type genotype (adjusted P values of <0.05), except those data points falling on 0 Arrows indicate the directions of changes in expression levels between transgenic mice.

Although no histologically distinct medullary region is detectable (Fig. 4B), the expression levels of genes indicative of different subsets of mTECs (Fig. 4C) are higher than in the Bl_Foxn4-driven epithelium. Although the morphology of the transgenic thymus lacks the characteristic zonation of cTECs and mTECs that is typical of the wild-type epithelium, low levels of Aire gene expression are detectable (Fig. 4C). Immunohistological analysis indicates the presence of only few Aire+ TECs, which are observed in the edges of epithelial cellfree regions in the reconstituted thymus, in contrast to the obvious Aire+ mTEC clusters in the wild-type thymus (Fig. 4D).

In line with the remarkable sequence conservation of vertebrate Foxn4 proteins (figs. S1, S2, and S5), the structures of the thymic microenvironments driven by Cm_Foxn4 (Fig. 4B) and Mm_Foxn4 (32) genes are essentially identical, despite 500 million years (Ma) of independent evolution. However, compared to the situation in Cm_Foxn4 transgenic mice, Mm_Foxn4-driven epithelia express lower levels of Prss16 and Aire, suggesting that with evolutionary progression, Foxn4 genes have gradually lost the capacity to support maturation of both cTECs and mTECs (Fig. 4E).

Previously, we have shown that the B cells generated in the Mm_Foxn4 thymus are preferentially located in close proximity to the vasculature, i.e., in the mesenchymal perivascular space (22). This finding highlighted the presence of anatomically distinct niches supporting the development of the two principal lymphocyte lineages in a primordial thymus; T cells differentiate in an epithelial environment, whereas B cells differentiate in a mesenchymal niche, similar to the situation in the sinusoidal environment of the bone marrow (33). In notable similarity, B cells in the Cm_Foxn4-expressing thymi are again found in the perivascular space (Fig. 4F), indicating that Foxn4 proteins favor the formation of anatomically separated domains specialized in either T or B cell differentiation.

Next, we tested the thymopoietic capacity of a Foxn1 gene that is suggested by phylogenetic analysis to have emerged from the ancestral vertebrate Foxn1/4-like gene, following a gene duplication event at the base of vertebrate evolution (Fig. 1 and figs. S1 and S2). With respect to mammalian Foxn1 genes, the elephant shark Cm_Foxn1 gene is the evolutionarily most distant form of a Foxn1-like gene of jawed vertebrates examined here (Fig. 1 and figs. S1, S2, and S6). Despite more than 400 Ma of independent evolution, the lymphopoietic capacity of shark Cm_Foxn1 is remarkably similar to that of mouse Mm_Foxn1. The total numbers of CD4+CD8+ immature thymocytes in Cm_Foxn1-expressing thymi approach those observed in mouse wild-type thymi; moreover, differentiation into CD4+ and CD8+ single-positive T cells occurs; as a result, the relative proportions of immature and mature thymocytes are identical to wild-type thymi (Fig. 3, B and D). With respect to intrathymic B cell development, we find that the absolute numbers of immature B cells are about 10-fold lower in the Cm_Foxn1 transgenic thymi than in the corresponding Cm_Foxn4 reconstitution (Fig. 3, C and E); however, they are slightly increased when compared to the wild-type mouse thymus.

As expected from the near-normal hematopoietic compartment in Cm_Foxn1 thymi, their microenvironment also resembles a wild-type mouse thymus. This is evident from the cytometric profiles of Ly51 and UEA1 markers (Fig. 4A) and the anatomical segregation of K5+K8 (medullary) and K5K8+ (cortical) areas in tissue sections (Fig. 4B); except for a somewhat smaller overall size, the histology of the Cm_Foxn1 thymus is essentially identical to the wild-type form. As expected, Aire+ TECs are present in the distinct medullary areas, sparing the thymic cortex (Fig. 4E). The expression patterns of signature genes in Cm_Foxn1 TECs indicate the remarkable similarity to wild-type TECs in the mouse thymus (Fig. 4G). However, subtle differences in the gene expression profiles exist; the increased levels of Prss16 suggest the presence of a bias toward mature cTECs at the expense of certain mTEC populations, for instance, those expressing Trpm5 (Fig. 4G). Nonetheless, our observations suggest that the T cellbiased lymphopoietic properties of Foxn1-like genes, as exemplified by the mammalian thymus, had already emerged in the ancestor of jawed vertebrates and were maintained throughout subsequent evolution.

With respect to the reconstructions described above, it is important to note that they represent an artificial disentanglement of Cm_Foxn1 and Cm_Foxn4 functions, since in the thymus of cartilaginous fish, Foxn1 and Foxn4 paralogs are coexpressed (fig. S7) (22). However, the expression patterns of the two genes are not completely identical; whereas they are both expressed in the shark retina, the expression of Foxn4 in distinct cell clusters in the spinal cord is unique (fig. S7). To mimic the physiological coexpression of both genes in TECs of cartilaginous fish, we generated Cm_Foxn1/Cm_Foxn4 double-transgenic mice (hereafter Cm_dtg). When compared to the number of thymocytes in Cm_Foxn4 and Cm_Foxn1 single-transgenic mice, this constellation of coexpression results in an intermediate number of T cells, although the cellularity is closer to the situation in Cm_Foxn1 thymi (Fig. 3A). Unexpectedly, coexpression of Cm_Foxn1 and Cm_Foxn4 leads to markedly increased numbers of immature B cells, almost two orders of magnitude more than in the Cm_Foxn1 single transgenic (Fig. 3, C and E). The higher capacity for B cell development in the double-transgenic thymus does not affect their anatomical localization; B cells still reside in close proximity to the vasculature, as seen in thymi driven by the expression of Cm_Foxn4 alone (Fig. 4F). The TECs of the bipotent lymphoid organ in double-transgenic mice exhibit a phenotype intermediate between the single transgenics, as reflected in their cell surface phenotype (Fig. 4A); cTECs expressing high levels of Ly51 still predominate, although UEA1+ mTECs make up a quarter of the TEC compartment. The keratin expression pattern indicates that the medullary and cortical regions are not as precisely demarcated as in the Cm_Foxn1 single transgenic (Fig. 4B); however, the medulla contains a large number of Aire+ cells (Fig. 4E), in line with the gene expression pattern (Fig. 4G). In terms of lymphopoietic capacity, the double-transgenic TECs are distinguished by a markedly increased capacity for B cell development (about 10 times higher than that of the Cm_Foxn4 thymus), whereas the capacity for immature T cells is only slightly reduced compared to the Cm_Foxn1 single-transgenic thymus (Fig. 3, G and H). The unique morphological and functional characteristics of the double-transgenic thymus are reflected in an intermediate gene expression pattern, demonstrating that the combination of signature genes selected here faithfully report the thymopoietic characteristics of TECs. Collectively, our results indicate that the coexpression of Cm_Foxn1 and Cm_Foxn4 in the same TECs recapitulates the observed bipotent nature of lymphopoiesis in the thymi of extant cartilaginous fishes (34), providing an important validation of the biological relevance of the present reconstruction strategy.

Whereas Foxn4 proteins are well conserved during the course of vertebrate evolution (fig. S5), Foxn1 proteins are much more variable (Fig. 1 and fig. S6) in particular, with respect to the lengths and amino acid sequence compositions of their N-terminal domains. When viewed through the lens of the mouse proteins, the sequences encoded in coding exons 2 and 3 of mouse Foxn4 are replaced by unrelated sequences in a single exon (coding exon 2) of mouse Foxn1 (fig. S8). This observation suggested that, after gene duplication, the evolution of protein domain(s) in Foxn1 proteins was associated with increasing lymphoid selectivity, from bipotency of Foxn4 to unipotency of Foxn1. We set out to test this hypothesis by creating a chimeric protein, Foxn1*4, in which the sequence encoded by coding exon 2 of the mouse Foxn1 gene was replaced by coding exons 2 and 3 of mouse Foxn4 (Fig. 5A and fig. S8). The overall cellularity in the thymus of Mm_Foxn1*4 transgenic mice was reduced to about 20% of wild-type numbers, but the differentiation of T cells occurred normally, as revealed by the presence of normal percentages of CD4+ and CD8+ single-positive cells (Fig. 5, B and C). However, the CD4CD8 double-negative compartment was increased in the Mm_Foxn1*4 transgenic thymi, likely caused by an increase in the number of B cells (Fig. 5D). Expression of the Mm_Foxn1*4 chimeric protein led to a 10-fold increase of B cell poietic capacity, without appreciable loss of T cell poietic capacity, when compared to Mm_Foxn1 (Fig. 5, E and F).

(A) Schematic representation of the N-terminal domains of mouse Foxn4, Foxn1, and the Foxn1*4 chimera; boxes correspond to exons, and colored lines correspond to conserved amino acid residues in Foxn4 and Foxn1 clades (see figs. S2 and S8 for details). The > sign denotes the DNA binding and activation domains, which are not shown here. (B) Intermediate cellularity of Foxn1*4 thymi (***P < 0.001; two-tailed t test); wt (n = 5), Mm_Foxn4 (n = 6), and Foxn1*4 (n = 13). (C) Enlarged CD4CD8 DN compartment in Foxn1*4 thymi (P < 0.001; two-tailed t test); wt (n = 5) and Foxn1*4 (n = 13). (D) Moderately increased numbers of IgMCD93+ immature B cells (P = 0.293; two-tailed t test, compared to wt); wt (n = 3) and Foxn1*4 (n = 7). (E) Foxn1*4 supports intrathymic T cell development (P = 0.6654; two-tailed t test, compared to wt); wt (n = 5) and Foxn1*4 (n = 13). (F) Increased B cell development (*P = 0.0335; two-tailed t test, compared to wt); wt (n = 8), Mm_Foxn4 (n = 7), and Foxn1*4 (n = 13). In (E) and (F), data for Mm_Foxn4 transgenics (shaded area) are taken from (22). (G) Flow cytometric analyses of Ly51 expression and UEA1 binding of EpCAM+CD45 TECs; wt (n = 5) and Foxn1*4 (n = 13). (H) Epithelial microenvironment of reconstructed thymi resolved by keratin 5 (K5) (in green) and K8 (in red) staining; 4-week-old mice. (I) Localization of B220+ B cells (in green) adjacent to ER-TR7+ mesenchyme (in red); inset shows a higher magnification of the indicated region highlighting the perivascular space. (J and K) Differential gene expression patterns in TECs; see legend in Fig. 4 for details; wt (n = 3), Mm_Foxn4 (n = 6), Foxn1*4 (n = 3), and Cm_dtg (n = 5). In (B), (E), and (F), each data point represents one mouse.

Cell surface markers (Fig. 5G) of TECs in the Mm_Foxn1*4 thymus are reminiscent of the reconstructed thymus in Cm_dtg mice (Fig. 4A). This is evident from a 10-fold greater fraction of Ly51+UEA1 cTECs in the transgenic thymus, with a corresponding reduction of Ly51UEA1+ mTECs. This conclusion is supported by the pattern of K5 and K8 keratin expression and the histological features of cortical and medullary areas (Fig. 5H). Notably, as is the case in Cm_dtg transgenic thymi (Fig. 4F), B cells are situated in the perivascular space of the thymus of Mm_Foxn1*4-transgenic mice (Fig. 5I). Collectively, this finding demonstrates that the sequences in the evolutionarily dynamic N-terminal domains of Foxn1 and Foxn4 proteins have important roles in controlling the extent of B cell development in the thymic microenvironment. In further support of this conclusion, we find that the lancelet Foxn4 protein [which does not support B cell development (Fig. 2, C, D, and I)] lacks most of the conserved amino acid sequence signature of the vertebrate Foxn4 family in this domain (fig. S2). Last, as expected from flow cytometric profiles of TECs and the distinct histological features seen in the tissue sections, the gene expression signature of Foxn1*4-expressing TECs suggests a bias toward the cTEC lineage at the expense of mature mTECs (Fig. 5J), much like the situation in Cm_dtg mice (Fig. 5K).

Our results provide a previously unattainable possibility to compare the transcriptional profiles of thymic microenvironments established by the activity of the cephalochordate Bl_Foxn4, the combinatorial activities of shark Cm_Foxn1 and Cm_Foxn4, and the activity of the mammalian Mm_Foxn1 genes. On the basis of the expression levels of selected TEC signature genes, a clear evolutionary trend becomes apparent: A gradual decrease in expression of cTEC-like genes is accompanied by an increase in expression of the genes that are characteristic of the mTEC compartment (Fig. 6A). This genetic constellation is associated with the more than 100-fold increase in overall T lymphopoietic capacity when comparing the thymic microenvironment established by the activities of the amphioxus Bl_Foxn4 and the mammalian Mm_Foxn1 genes. At the same time, appreciable B lymphopoietic capacity appears to have been a transient phenomenon, absent in Bl_Foxn4-driven and Mm_Foxn1-driven thymic microenvironments but present in the unique Foxn1 and Foxn4 coexpressing microenvironments characteristic of cartilaginous and teleost fishes (22). Hence, we became interested to determine the potential mechanistic basis of the bipotent microenvironment. Our previous experiments (22) suggested that the ratio of Dll4 and Il7 expression levels in the thymic microenvironment is an important determinant of its lymphopoietic properties, with respect to the balance between T and B cell poiesis. The Notch1/Dll4 signaling pathway is essential for the initiation of T cell development in the thymus (5, 15, 16, 35), whereas Il7 functions as a general lymphopoietic growth factor (36). The Dll4 gene is known to be a direct target of the Foxn1 (5, 37) and Foxn4 (4, 38) transcription factors, whereas the expression of Il7 is independent of Foxn1 activity (39). In support of the latter conclusion, we observed similar expression levels of the Il7 gene in all reconstituted TEC compartments examined here. Hence, since the denominator in the Dll4/Il7 ratio is a constant, it follows that the Foxn1/4-dependent expression levels of Dll4 determine the particular type of lymphopoietic activity in the thymus. In the reconstructions with Cm_Foxn1 and Cm_Foxn4 genes, we observed that, compared to the single transgenics, the Cm_dtg thymus had the highest capacity for B cell development (Fig. 6B), in line with the natural bipotency of the thymus of cartilaginous fish (34, 40). The ratio of T cell poiesis and B cell poiesis (here referred to as the T/B index) positively correlated with the ratio of expression levels of Dll4 and Il7 genes (Fig. 6C). In the comparison of Mm_Foxn4, Mm_Foxn1, and Mm_Foxn1*4 transgenics, the same positive relationship between T/B index and Dll4/Il7 ratio holds (Fig. 6, D and E). As compared to the situation in wild-type mice, Dll4 expression is several fold higher in Bl_Foxn4 transgenic microenvironments than it is in Cm_Foxn4-driven TECs, reflecting the T cell bias in the former and the presence of B cell development in the latter. Overall, the Dll4 expression levels vary by about one order of magnitude (Figs. 4, C, E, and G, and 5, J and K). Although it is likely that other factors affect the lymphopoietic properties of the thymic microenvironment, the modulation of Dll4 expression through Foxn1/4 transcription factors emerges as an evolutionarily conserved and functionally relevant mechanism by which the lymphopoietic capacity and the bias for or against B cell development of the thymus could be modulated.

(A) Differential gene expression patterns in TECs; see legend in Fig. 2 for details. (B and D) T and B cell poietic indices; arrows indicate the altered balance between T and B cell generation. (C and E) Ratios of T and B cell indices as a function of the ratios of Dll4 and Il7 expression levels; Cm_Foxn4 (n = 6), Cm_Foxn1 (n = 7), Cm_dtg (n = 5), wt (n = 3), Mm_Foxn4 (n = 6), and Foxn1*4 (n = 7). ***P < 0.001 and *P < 0.05; one-way ANOVA with Tukeys multiple comparison test; SDs correspond to propagated errors. (F) Schematic summarizing gene content and expression characteristics and associated lymphopoietic properties of thymi during vertebrate evolution.

The in vivo reconstitution experiments described here suggest a sequence of events during vertebrate evolution that culminated in the emergence of the T cellbiased thymus. We hypothesize that the primordial vertebrate Foxn4-like gene was expressed in the pharyngeal endoderm in the ancestor common to all vertebrates, much like its ortholog in lancelets (4). After the emergence of lymphocytes, which may have had their evolutionary origin in lymphocyte-like cells of tunicates (41), the Foxn4-expressing patch of pharyngeal endoderm cells may have supported the development of T-like cells; we further propose that this primordial type of lymphopoietic activity initially supported their development only up to the stage where their germline-encoded antigen receptors were expressed (Fig. 6F). Our previous studies demonstrated that the expression of the Notch ligand Dll4 and the chemokine Cxcl12 in Foxn1-deficient TECs suffices to support T cell differentiation up to the CD4+CD8+ DP stage (5), albeit at a much lower efficiency than we observe here in Bl_Foxn4 reconstitutions. Nonetheless, these results collectively suggest that a small number of effector molecules suffice to jump-start the formation of a lymphopoietic environment. We consider it likely that at this particular stage of immune system evolution, the facility of somatic recombination of antigen receptor genes and an associated quality control mechanism(s) mitigating any potential autoreactivity was not yet established (42). Nonetheless, lymphocytes in the vertebrate ancestors immune system may have expressed different kinds of germline-encoded antigen-specific receptors, analogous to pattern recognition receptors; variegated expression of these sensory modules would have afforded early vertebrates with the capability of immune responses and memory functions through clonal proliferation, akin to natural killer cells in mammals (43). Collectively, the phenotype of the Bl_Foxn4 epithelium defines a previously unidentified checkpoint during TEC differentiation, which marks the support of T cell development up to the CD4+CD8+ DP stage.

The emergence of a typical vertebrate-like Foxn4 gene (here exemplified by Cm_Foxn4) heralds another critical transition point in the immune systems of early vertebrates, as it established an environment supporting the development of the two principal lymphocyte lineages. Within the epithelial TEC compartment, it fostered the further development of T cells to reach the single-positive stage, albeit at low efficiency, and at the interface between epithelial and mesenchymal components of the microenvironment, it established conditions conducive to B cell development, as indicated by the presence of substantial numbers of immature B cells in and around the perivascular space. A comparison of protein sequences suggests that changes in the N-terminal domain of Bl_Foxn4 facilitated this process. Since the predicted Foxn4 protein of tunicates assumes an intermediate position in the phylogeny of chordate Foxn1/4 proteins (Fig. 1), it will be of interest to examine its capacity to support the development of B cells.

After the emergence of the Foxn1 gene in a primordial vertebrate, as a result of a gene duplication event, coexpression in the pharyngeal epithelium of the paralogous Foxn4 and Foxn1 genes was maintained; this coexpression signature still persists in extant cartilaginous (fig. S7) (4) and bony fishes (22, 44). Protein sequence comparisons indicate that the emergence of the Foxn1 paralog was accompanied by a radical modification of the amino acid sequence composition of the N terminus. This finding strongly supports the notion that exon replacement event(s) accompanied the emergence of the first Foxn1 genes that exchanged two exons of the Foxn4 gene by a single exon of substantially different sequence. In the Cm_Foxn1 protein, the N terminus is much shorter than it is in the living representatives of evolutionarily more recent taxa, such as the mouse Foxn1 protein, suggesting that structural features of this domain rather than primary amino acid sequence similarities underlie the equivalent functionalities of the Cm_Foxn1 and Mm_Foxn1 proteins.

Because the overall number of TECs present in the thymic lobes was essentially invariant in all reconstructed thymi and similar in magnitude to that in the wild-type mouse thymus, the main difference between Bl_Foxn4 and the vertebrate versions of Foxn1/4 proteins is the much lower lymphopoietic capacity of the former. This observation indicates that the key functions of the Foxn1/4 proteins are to qualitatively alter epithelial cell phenotypes rather than acting to simply quantitatively expand the epithelial compartment via proliferation. Our results indicate that the increase in lymphopoietic capacity occurred in a stepwise fashion, from Bl_Foxn4 to Cm_Foxn4 to CmFoxn1. In addition, the transition from Foxn4 to Foxn1 was accompanied by an increased size of the mTEC compartment, associated with a larger proportion of mature single-positive T cells among thymocytes. In the embryonic mouse thymus, cTECs develop earlier than mTECs (45, 46); this ontogenetic sequence closely resembles the phenotypes we observe in the phylogenetic sequence of Bl_Foxn4 to Cm_Foxn4 to Cm_Foxn1.

Another critical transition in the evolutionary trajectory of Foxn1/4 genes and vertebrate thymopoiesis occurred when cis-regulatory changes led to the loss of Foxn4 expression in TECs. This reorganization of genetic networks is exemplified by the reciprocal expression patterns of Foxn1 and Foxn4 in chicken tissues (fig. S9). As a result, T cell development became entirely dependent on Foxn1 (9, 47). At present, we do not know why a degenerate network structure [note that Foxn4 and Foxn1 are partially redundant in the teleost thymus (22)] was replaced by a nonredundant design. However, since the contribution of the thymus to B cell development was abolished in this process, loss of Foxn4 expression in TECs helped establish the strict anatomical segregation of developing lymphocyte lineages (48). As a result, the thymus was eventually transformed into an organ highly specialized for efficient T cell development (Fig. 6F).

C57BL/6J mice are maintained in the Max Planck Institute of Immunobiology and Epigenetics. Foxn1:Bl_Foxn4, Foxn1:Cm_Foxn4, and Foxn1:Cm_Foxn1 transgenic mice were constructed according to standard protocols by cloning a 27,970base pair mouse Foxn1 promoter fragment (GenBank accession number Y12488; nucleotides 5680 to 33,650) upstream of either B. lanceolatum Foxn4 cDNA (GenBank accession number AJ252025.1; nucleotides 1 to 1590), C. milii Foxn4 cDNA (GenBank accession number FJ176202.1, nucleotides 56 to 1615), or a C. milii Foxn1 cDNA (GenBank accession number FJ176201.1; nucleotides 76 to 1584), followed by the bovine growth hormone polyadenylation sequence (32); cDNAs of Cm_Foxn4 and Cm_Foxn1 were synthesized by Eurofins Genomics and cloned into peX-K248 vector; in the Foxn1:Foxn1*4 construct, nucleotides 267 to 728 of M. musculus Foxn1 (GenBank accession number NM_008238.2) were replaced by nucleotides 170 to 445 of M. musculus Foxn4 (GenBank accession number AF323488.1). To generate transgenic mice, constructs were linearized and injected into FVB pronuclei according to standard protocols.

Transgenic mice were subsequently backcrossed to Foxn1-deficient mice (10) on a C57BL/6J background. Mice were kept in the animal facility of the Max Planck Institute of Immunobiology and Epigenetics under specific pathogenfree conditions. All animal experiments were performed in accordance with the relevant guidelines and regulations, approved by the review committee of the Max Planck Institute of Immunobiology and Epigenetics and the Regierungsprsidium Freiburg, Germany (license AZ 35-9185.81/G-14/57). Transgene expression levels were determined by RNA-seq, comparing transgene-derived BGH_PolyA transcripts to lacZ transcripts, the latter representing the activity of the targeted endogenous Foxn1 locus (10); the ratios of lacZ transcript counts to those emanating from the 5 part of the Foxn1 gene [note that the 5 end of the Mm-Foxn1 sequence is still detectable in the transcriptome of Foxn1/ mice (10)] served as normalization. A negative feedback loop suppresses the endogenous Foxn1 locus activity for transgenes encoding proteins functionally equivalent to Foxn1; despite a 10-fold difference in expression levels, the transgenes are expressed in the range of the endogenous Foxn1 gene in wild-type mice (fig. S10). For identification of transgenes, the following primers were used for genotyping: Foxn1:Mm_Foxn1, SS40 (wild-type allele) + SS35 (wild-type and knockout allele) + JBS003 (knockout allele), Foxn1:Mm_Foxn1*4, XAH388 + SS6; Foxn1:Cm_Foxn1, XAH163 + RM19; Foxn1:Cm_Foxn4, XAH163 + RM22; Foxn1:Bl_Foxn4, JBS465 + JBS466; SS40, 5-CTGTGAACTCAGCCATACTC; SS35, 5-TGCACCAAGCCTCTGCTGGGA; JBS003, 5-TCGCCTTCTTGACGAGTTCT; XAH388, 5-CAGCAACTGATAAGGTCACC; SS6, 5-ACAGAATTCTTCCAGCCATCA; XAH163, 5-GTCCCTAATCCGATGGCTAGCTC; RM19, 5-TATCGCGTGCACGAGTTGTA; RM22, 5-GGTTAAAGTTCATGCGGCCG; JBS465, 5-CCAGCTCCGAAACAGCCTAA; JBS466, 5-GTCCTTTGTCGTCTGGTCGT.

Thymus organs were fixed for 120 min in 4% paraformaldehyde, washed with phosphate-buffered saline (PBS), and incubated in 20% sucrose overnight before mounting and snap-freezing in optimal cutting temperature (OCT) embedding compound. Tissue sections (8 m) were cut using a cryostat and mounted onto precoated slides (Superfrost plus, Thermo Fisher Scientific). Slides were dried, followed by a 30-min blocking step using mouse immunoglobulin G (IgG) at 1:50 diluted in PBS + 0.5% bovine serum albumin (BSA) + 0.2% Tween. K5 K8 staining was performed with rabbit anti-K5 antibody (Ab) (PRB-160P, Covance) at 1:500 and rat anti-K8 Ab (Troma1, in-house) at 1:200. As secondary Ab, goat anti-rabbit Alexa Fluor 488 (A11008, Thermo Fisher Scientific) at 1:500 and donkey anti-rat Cy3 (AB_2340668, Jackson ImmunoResearch) at 1:500 were used. For ER-TR7 B220 staining, the rat antiER-TR7 Alexa Fluor 647 Ab (sc-73355 AF647, Santa Cruz Biotechnology) at 1:50 and rat anti-B220 Alexa Fluor 488 Ab (RA3-6B2, eBioscience) at 1:200 were used. Sections were mounted with Fluoromount G before analysis (Apotome, Zeiss). For combined K5/K8/Aire staining, sections were dried, blocked, and stained with unlabeled primary Abs as above and then stained with the secondary Abs donkey anti-rabbit IgG Cy3 (Jackson ImmunoResearch 711-165-152) and mouse anti-rat light chain Alexa Fluor 647 (MAR 18.5.28, purified and labeled in-house). Sections were then blocked with rat IgG and subsequently stained with rat anti-mouse Aire Alexa Fluor 488 Ab (5H12, eBioscience 14-5934-82) at 1:200.

RNA in situ hybridization was carried out essentially as described (22) using the following probes: Gallus gallus Foxn1, nucleotides 1371 to 2557 in GenBank accession number XM_415816.6; G. gallus Foxn4, nucleotides 559 to 1779 in GenBank accession number NM_001083359.1; Scyliorhinus canicula Foxn1, nucleotides 37 to 346 in GenBank accession number FJ187748; and S. canicula Foxn4, nucleotides 1 to 422 combined from GenBank accession numbers Y11538, Y11539, and Y11540, respectively.

To generate single-cell suspensions for analytical and preparative flow cytometry of TECs, the procedures in (49) were followed. Note that the enzymatic cocktail required to liberate TECs destroys the extracellular domains of CD4 and CD8 surface markers (but not that of the CD45 molecule); hence, when analysis of thymocyte subsets was desired, thymocyte suspensions were prepared in parallel by mechanical liberation, best achieved by gently pressing thymic lobes through 40-m sieves. Cell surface staining [anti-CD45 (30-F11), conjugated with phycoerythrin (PE) Cy7 (BioLegend); anti-EpCAM (G8.8), conjugated with allophycocyanin (APC; BioLegend); anti-Ly51 (BP-1) (6C3), conjugated with PE (eBioscience); UEA1, conjugated with fluorescein isothiocyanate (FITC; Vector Biosciences); anti-CD4 (GK1.5), conjugated with FITC (BioLegend); anti-CD8a (53-6.7), conjugated with APC (eBioscience); anti-CD19 (eBio1D3), conjugated with PerCPCy5.5 or PeCy7 (eBioscence); anti-B220 (CD45R) (RA3-6B2), conjugated with biotin (eBioscience); anti-IgM (II/4.1), conjugated with PE (eBioscience), anti-CD93 (C1qRp) (AA4.1), conjugated with APC (eBioscience); streptavidin conjugated with eFluor 450 or FITC (eBioscience)] was performed at 4C in PBS supplemented with 0.5% BSA and 0.02% NaN3. Because of their small size in Bl_Foxn4 mice, the numbers of TECs and hematopoietic cells in thymi were determined independently; hence, B and T poietic indices were calculated from mean values with error propagation.

Single-cell suspensions were prepared by TEC digest as described above. CD45EpCAM+ cells [negative enrichment using anti-CD45 magnetic-activated cell sorting (MACS) beads and antiTer-119 MACS beads, Miltenyi Biotec] were sorted directly into TRI reagent (T9424, Sigma-Aldrich). RNA isolation was performed according to standard protocols. Libraries were prepared using the Ultra RNA Library Prep Kit (Illumina). Samples were run on HiSeq2500 and sequenced to a depth of > 60 106 to 100 106 reads per sample.

The relevant Foxn1 and Foxn4 amino acid sequences can be found under the following GenBank accession numbers: Foxn1: Rhincodon typus (XM_020525471); C. milii (XM_007896499); Amblyraja radiata (XM_033046380); Erpetoichthys calabaricus (XM_028808185); Acipenser ruthenus (XM_034049625); Lepisosteus oculatus (XM_015367325); Danio rerio (XM_009291615); Microcaecilia unicolor (XM_030186024.1); Nanorana parkeri (XM_018555008.1); Xenopus laevis (XM_018248776.1); Xenopus tropicalis (XM_018091796); Podarcis muralis (XM_028708697); Geotrypetes seraphini (XM_033921305); Anolis carolinensis (XM_016997340); G. gallus (XM_415816); Corvus cornix cornix (XM_019287554); Monodelphis domestica (XM_001375795); Ornithorhynchus anatinus (XM_029082501); M. musculus (NM_008238); Homo sapiens (NM_001369369); Foxn4: B. lanceolatum (AJ252025); Branchiostoma belcheri (XP_019621093); Phallusia mammillata (LR785254); R. typus (XM_020536376); A. radiata (XM_033043787); C. milii (NM_001292643); L. oculatus (XM_006640210); E. calabaricus (XM_028824927); D. rerio (NM_131099); Latimeria chalumnae (XM_014493627); Rhinatrema bivittatum (XM_029571604); X. laevis (BC142562); X. tropicalis (NM_001102862); G. seraphini (XM_033957269); P. muralis (XM_028705153); G. gallus (NM_001083359); C. cornix cornix (XM_020584559); Phascolarctos cinereus (XM_021008024); M. musculus (NM_148935); H. sapiens (NM_213596). The sequence of Foxn4 of Ciona intestinalis has been retrieved from the ENSEMBL database (ENSCING00000017653). Sequences were aligned using multiple sequence comparison by log-expectation (MUSCLE) (50), and the phylogenetic tree was reconstructed using the neighbor-joining method implemented in the BioNJ software (51) with 1000 bootstrap replicates and the Jones-Taylor-Thornton (JTT) substitution model (52). Both programs are available on the platform phylogeny.fr (53). The resulting phylogeny was visualized using the Interactive Tree of Life platform (54).

Transcriptomes were analyzed on the Galaxy platform using featureCounts (55) followed by DESeq2 analysis (56).

t tests (two-tailed) were used to determine the significance levels of the differences between the means of two independent samples, considering equal or unequal variances as determined by the F test. For multiple tests, the conservative Bonferroni correction was applied or as indicated, using one-way analysis of variance (ANOVA) with Tukeys multiple comparison test.

Excerpt from:
Retracing the evolutionary emergence of thymopoiesis - Science Advances

Read More...

Exploring First-line Therapies in Peripheral T-Cell Lymphomas – Targeted Oncology

Tuesday, November 24th, 2020

During the Targeted Oncology Case Based Peer Perspectives event, Javier L. Munoz, MD, Hematologist/Oncologist Director, Lymphoma Program Mayo Clinic, discussed the case of 60-year-old patient with peripheral T-cell lymphoma (PTCL).

Targeted Oncology: What are the standard regimens in treating T-cell lymphoma?

MUNOZ: Historically, we have been prescribing CHOP [cyclophosphamide, doxorubicin hydrochloride, vincristine sulfate, and prednisone] or CHOP-like regimens. There are retrospective German data showing the addition of etoposides, so CHOEP instead of CHOP, may be beneficial, particularly for patients who were younger than 65 years.1 That is not a randomized trial, it was a retrospective study, so the strength of the data was relatively weak.1 The data of brentuximab vedotin [Adcetris] plus CHP [cyclophosphamide, doxorubicin, prednisone] versus CHOP [was from] a randomized trial, so its a stronger level of evidence compared with that etoposide data.

What do the National Comprehensive Cancer Network (NCCN) guidelines recommend?

The category 1 recommendation for ALCL [anaplastic large cell lymphoma] is brentuximab vedotin plus CHP, according to the NCCN.2 For that particular indication, I think most people are not going to argue that brentuximab plus CHP is the way to go.

For the other subtypesand numbers for the other subtypes were smaller compared with ALCLits a bit more blurry. You still have brentuximab vedotin plus CHP, but you also have the options of CHOP, CHOEP.

What was the rationale for ECHELON-2 [NCT01777152] in this population? This was a randomized study, brentuximab vedotin plus CHP versus CHOP.3 The bottom line is that this trial was a replacement strategy. Vincristine, or Oncovin, is a vinca alkaloid, and it causes neuropathy. For brentuximab, an antibody-drug conjugate, its payload is also a vinca drug. Vinca alkaloids cause neuropathy, so the trial replaced those agents, resulting in this trial. It was a double-dummy, [double-blind] study, so patients received placebobrentuximab and CHP or they received CHOP. The CD30 expression needed to be 10% or higher for the patient to be able to enroll. The investigators allowed multiple subtypes of T-cell lymphomas: PTCL not otherwise specified, angioimmunoblastic T-cell lymphoma, hepatosplenic T-cell lymphoma, enteropathy-associated T-cell lymphoma.

The reality is that most of you are going see PTCL, not otherwise specified, because its the most common subtype I see; maybe the runner-up is angioimmunoblastic T-cell lymphoma, and then the next one is going to be ALCL. And again, there are a couple of flavors: ALK-positive and ALK-negative. ALK-negative is, prognostically, closer to PTCL. The primary end point was progression-free survival [PFS] but, of course, they looked at other things like overall survival.3

What were the outcomes of this trial for patients? PFS was a primary end point, and this was the most important factor or variable that they looked at, and with CHOP you get 20.8 months of PFS; with brentuximab plus CHP you got 48.2 months. So, more than double the primary end point, and that is why I like to start with PTCL, because ECHELON-2 definitely looked more impressive than the ECHELON-1 trial [NCT01712490]. Theyre both important papers; ECHELON-1 was published in New England Journal of Medicine,4 ECHELON-2 was only published in Lancet. The difference, the numbers that you see, are more dramatic in ECHELON-2 in PTCL compared with Hodgkin lymphoma, but both are relevant papers. More than double the results, and you could see that that separation is maintained over time.

They looked at overall survival, and there was also a difference. Even though the primary end point was PFS, your patients are also going to live longer if you go with brentuximab plus CHP.

The complete response rate was 68% for [brentuximab vedotin plus CHP] versus 56% for CHOP. Of course, the objective response rate is also higher for brentuximab plus CHP, as well [TABLE5 ].

As I said, both drugs can potentially cause neuropathy. Lets take a look at the safety profile. Peripheral neuropathy for brentuximab plus CHP, grade 3 or higher, was 4%, for CHOP, it was 3%. Remember, vincristine in the CHOP regimen can also cause neuropathy. Not a big difference there. For any grade of neuropathy, the numbers are 45% for brentuximab plus CHP and 41% for CHOP.

Now, could you die from CHOP, or brentuximab plus CHP? Of course. Luckily for us, its single digits. Its rareIm sure that we have all had patients that we have lost during CHOP or R-CHOP [rituximab (Rituxan) plus CHOP] for B-cell lymphomas, but 3% for brentuximab plus CHP and 4% for CHOP.

If this patient achieved a complete response to frontline therapy, would you consider transplant?

Before the ECHELON trials, most of us knew that CHOP was suboptimal; we just did not know what was better than CHOP. Thats why we were playing with etoposide, based on that retrospective German data and doing CHOEP, sometimes even EPOCH [etoposide phosphate, prednisone, vincristine sulfate, cyclophosphamide, and doxorubicin hydrochloride] for these patients. Because CHOP was suboptimal, we wanted to do something additional, and that is why many patients have received consolidation with transplants.

We consider so many factors for transplant-eligible patientsthe performance status, comorbidities, age; but in general, Im prone to offer [transplant] to patients who have these T-cell lymphomas because, again, its a bad disease, and its not a curative intent. You are not giving the treatment with a curative intent. You want to keep the disease at bay as much as possible.

What is the impact of consolidative stem cell transplant after frontline brentuximab plus CHP for patients?

In a post hoc analysis, for the patients who achieved complete remission [CR], 38 patients in CR, received the transplant, and 76 patients in CR did not; it was certainly not the primary end point.5 It was presented at the American Society of Hematology Annual Meeting in 2019. Again, we are tempted to prescribe the transplant because, historically, these patients have done so poorly. The majority of these patients in CR after brentuximab plus CHP did not receive a transplant, 38 patients did receive a transplant, and it seems that the patients that received the consolidation with the transplant did better than the patients who did not receive the transplant. You have the 3-year progression-free survival numbers: 76% and 53%, though my bias at this point would be young, relatively young, or with few comorbidities, good performance statusI would certainly send the patient to get a second opinion.

What would be your approach toward second-line therapy at progression?

The number 1 option is a clinical trial. Id also consider pralatrexate [Folotyn]. Belinostat [Beleodaq], pralatrexate, and romidepsin [Istodax] are going to give an overall response somewhere between 20% to 30%, and complete remissions that are approximately 10%, give or take. Duration of response is usually going to be less than a year. Bottom line, we need to do better for PTCL when its relapsed/refractory. We need to do good when it comes to our frontline therapies, so, hopefully, we can provide a long period of time of remission for our patients.

References:

1. Schmitz N, Trmper L, Ziepert M, et al. Treatment and prognosis of mature T-cell and NK-cell lymphoma: an analysis of patients with T-cell lymphoma treated in studies of the German high-grade non-Hodgkin lymphoma study group. Blood. 2010;116(18):3418- 3425. doi:10.1182/blood-2010-02-270785

2. NCCN. Clinical practice guidelines in oncology. T-cell lymphomas, version 1.2021. Accessed October 14, 2020. https://bit.ly/3kcUHLL

3. Horwitz S, OConnor OA, Pro B, et al; ECHELON-2 Study Group. Brentuximab vedotin with chemotherapy for CD30-positive peripheral T-cell lymphoma (ECHELON-2): a global, double-blind, randomised, phase 3 trial. Lancet. 2019;393(10168):229-240. doi:10.1016/S0140-6736(18)32984-2

4. Connors JM, Jurczak W, Straus DJ; ECHELON-1 Study Group. Brentuximab vedotin with chemotherapy for stage III or IV hodgkins lymphoma. N Engl J Med. 2018;378(4):331-344. doi: 10.1056/NEJMoa1708984

5. Savage KJ, Horwitz SM, Advani RH. An exploratory analysis of brentuximab vedotin plus chp (A+CHP) in the frontline treatment of patients with CD30+ peripheral T-cell lymphomas (ECHELON-2): impact of consolidative stem cell transplant. Blood. 2019; 134(suppl 1):464. doi:10.1182/blood-2019-122781

Link:
Exploring First-line Therapies in Peripheral T-Cell Lymphomas - Targeted Oncology

Read More...

Adaptive Biotechnologies and Collaborators to Present Data from More Than 35 Abstracts at ASH 2020 Highlighting Clinical Relevance of MRD Testing with…

Tuesday, November 24th, 2020

SEATTLE, Nov. 23, 2020 (GLOBE NEWSWIRE) -- Adaptive Biotechnologies Corporation (Nasdaq: ADPT), a commercial stage biotechnology company that aims to translate the genetics of the adaptive immune system into clinical products to diagnose and treat disease, together with its collaborators will present data from more than 35 abstracts studying the use of Adaptives clonoSEQ Assay for minimal residual disease (MRD) assessment at the American Society of Hematology (ASH) virtual 62nd Annual Meeting and Exposition, December 5-8. clonoSEQ is the first and only U.S. Food and Drug Administration (FDA)-cleared assay for MRD assessment in chronic lymphocytic leukemia (CLL), multiple myeloma and B-cell acute lymphoblastic leukemia (B-ALL) and is widely available to clinicians and patients across the U.S.

We are thrilled to see so many investigators presenting clonoSEQ data at ASH this year, among the more than 300 ASH studies highlighting MRD data, significantly growing the body of evidence validating this tool as a critical measure of patient outcomes, said Lance Baldo, MD, Chief Medical Officer of Adaptive Biotechnologies. As innovation continues for the treatment of blood cancers with novel and highly targeted therapies that create deep and durable responses for patients, we see clinicians increasingly utilizing clonoSEQ to help guide day-to-day patient care.

Assessment of MRD is a way to directly detect and quantify remaining disease, even in the absence of symptoms, across a spectrum of blood cancers. A patients MRD status gives clinicians timely information about how a patient may be responding to treatment, so patients and providers can be in control when it comes to managing their disease and treatment decisions.

clonoSEQ, the first clinical application of Adaptives immune medicine platform, will be featured in 14 oral presentations and 23 posters at ASH. Data on clinical and research utility from studies, as well as findings based on real-world experience, will be presented across a range of cancers including multiple myeloma, ALL, CLL and non-Hodgkins lymphoma (NHL). These new data show a correlation between clonoSEQ MRD results and improved blood cancer patient outcomes, enhanced clinical decision-making, and potential savings to the healthcare system.

Additional data at ASH this year will highlight Adaptives immune profiling research tool, immunoSEQ, to quantitatively assess the immune response to novel therapies in development.

Key presentations include:

AbouttheclonoSEQ AssayThe clonoSEQ Assay is the first and only FDA-cleared assay for MRD in chronic lymphocytic leukemia (CLL), multiple myeloma (MM) and B-cell acute lymphoblastic leukemia (ALL). Minimal residual disease (MRD) refers to the small number of cancer cells that can stay in the body during and after treatment. clonoSEQ was initially granted De Novo designation and marketing authorization by the FDA for the detection and monitoring of MRD in patients with MM and B-ALL using DNA from bone marrow samples.InAugust 2020, clonoSEQ received additional clearance from theFDA to detect and monitor MRD in blood or bone marrow from patients with CLL.

The clonoSEQ Assay leverages Adaptives proprietary immune medicine platform to identify and quantify specific DNA sequences found in malignant cells, allowing clinicians to assess and monitor MRD during and after treatment. The assay provides standardized, accurate and sensitive measurement of MRD that allows physicians to predict patient outcomes, assess response to therapy over time, monitor patients during remission and predict potential relapse. Clinical practice guidelines in hematological malignancies recognize that MRD status is a reliable indicator of clinical outcomes and response to therapy, and clinical outcomes have been shown to be strongly associated with MRD levels measured by the clonoSEQ Assay in patients diagnosed with CLL, MM and ALL.

The clonoSEQ Assay is a single-site test performed at Adaptive Biotechnologies.In addition to its FDA-cleared uses, clonoSEQ is also available as a CLIA-validated laboratory developed test (LDT) service for use in other lymphoid cancers and sample types. For important information about the FDA-cleared uses of clonoSEQ, including the full intended use, limitations, and detailed performance characteristics, please visitwww.clonoSEQ.com/technical-summary.

About immunoSEQ Assay Adaptives immunoSEQ Assay helps researchers make discoveries in areas such as oncology, autoimmune disorders, infectious diseases and basic immunology. The immunoSEQ Assay can identify millions of T- and B-cell receptors from a single sample in exquisite detail. Offered as a Service or Kit, the immunoSEQ Assay is used to ask and answer translational research questions and discover new prognostic and diagnostic signals in clinical trials. The immunoSEQ Assay provides quantitative, reproducible sequencing results along with access to powerful, easy-to-use analysis tools. The immunoSEQ Assay is for research use only and is not for use in diagnostic procedures.

About AdaptiveAdaptive Biotechnologies is a commercial-stage biotechnology company focused on harnessing the inherent biology of the adaptive immune system to transform the diagnosis and treatment of disease. We believe the adaptive immune system is natures most finely tuned diagnostic and therapeutic for most diseases, but the inability to decode it has prevented the medical community from fully leveraging its capabilities. Our proprietary immune medicine platform reveals and translates the massive genetics of the adaptive immune system with scale, precision and speed to develop products in life sciences research, clinical diagnostics and drug discovery. We have two commercial products and a robust clinical pipeline to diagnose, monitor and enable the treatment of diseases such as cancer, autoimmune conditions and infectious diseases. Our goal is to develop and commercialize immune-driven clinical products tailored to each individual patient.

For more information, please visit adaptivebiotech.com and follow us on http://www.twitter.com/adaptivebiotech.

Forward Looking Statements This press release contains forward-looking statements that are based on managements beliefs and assumptions and on information currently available to management. All statements contained in this release other than statements of historical fact are forward-looking statements, including statements regarding our ability to develop, commercialize and achieve market acceptance of our current and planned products and services, our research and development efforts, and other matters regarding our business strategies, use of capital, results of operations and financial position, and plans and objectives for future operations.

In some cases, you can identify forward-looking statements by the words may, will, could, would, should, expect, intend, plan, anticipate, believe, estimate, predict, project, potential, continue, ongoing or the negative of these terms or other comparable terminology, although not all forward-looking statements contain these words. These statements involve risks, uncertainties and other factors that may cause actual results, levels of activity, performance or achievements to be materially different from the information expressed or implied by these forward-looking statements. These risks, uncertainties and other factors are described under "Risk Factors," "Management's Discussion and Analysis of Financial Condition and Results of Operations" and elsewhere in the documents we file with theSecurities and Exchange Commissionfrom time to time. We caution you that forward-looking statements are based on a combination of facts and factors currently known by us and our projections of the future, about which we cannot be certain. As a result, the forward-looking statements may not prove to be accurate. The forward-looking statements in this press release represent our views as of the date hereof. We undertake no obligation to update any forward-looking statements for any reason, except as required by law.

MEDIA CONTACT:Beth Keshishian917-912-7195media@adaptivebiotech.com

ADAPTIVE INVESTORS:Karina Calzadilla201-396-1687

Carrie Mendivil, Gilmartin Groupinvestors@adaptivebiotech.com

Read the original post:
Adaptive Biotechnologies and Collaborators to Present Data from More Than 35 Abstracts at ASH 2020 Highlighting Clinical Relevance of MRD Testing with...

Read More...

Circulating Tumor Cells (CTC) Market Growth Analysis and Forecast to 2026 by Applications and Competitors – Cheshire Media

Tuesday, November 24th, 2020

The Circulating Tumor Cells (CTC) Market was valued at US$ XX million in 2019 and is projected to reach US$ XX million by 2025, at a CAGR of XX percentage during the forecast period. In this study, 2019 has been considered as the base and 2020 to 2025 as the forecast period to estimate the market size for Circulating Tumor Cells (CTC) Market

Deep analysis about market status (2016-2019), competition pattern, advantages and disadvantages of products, industry development trends (2019-2025), regional industrial layout characteristics and macroeconomic policies, industrial policy has also been included. From raw materials to downstream buyers of this industry have been analysed scientifically. This report will help you to establish comprehensive overview of the Circulating Tumor Cells (CTC) Market

Get a Sample Copy of the Report at: https://i2iresearch.com/report/global-circulating-tumor-cells-(ctc)-market-2020-market-size-share-growth-trends-forecast-2025/#download-sample

The Circulating Tumor Cells (CTC) Market is analysed based on product types, major applications and key players

Key product type:Ex-Vivo Positive SelectionIn-Vivo Positive SelectionNegative SelectionMicrochips & Single Spiral Micro Channel

Key applications:Tumorigenesis ResearchEmt Biomarkers DevelopmentCancer Stem Cell ResearchOthers

Key players or companies covered are:AdnaGenApocellBiocepCanopus BioscienceCreatv MicrotechIkonisysIV DiagnosticsMiltenyi BiotechNanostring TechnologiesRarecells DiagnosticsVitatex

The report provides analysis & data at a regional level (North America, Europe, Asia Pacific, Middle East & Africa , Rest of the world) & Country level (13 key countries The U.S, Canada, Germany, France, UK, Italy, China, Japan, India, Middle East, Africa, South America)

Inquire or share your questions, if any: https://i2iresearch.com/report/global-circulating-tumor-cells-(ctc)-market-2020-market-size-share-growth-trends-forecast-2025/

Key questions answered in the report:1. What is the current size of the Circulating Tumor Cells (CTC) Market, at a global, regional & country level?2. How is the market segmented, who are the key end user segments?3. What are the key drivers, challenges & trends that is likely to impact businesses in the Circulating Tumor Cells (CTC) Market?4. What is the likely market forecast & how will be Circulating Tumor Cells (CTC) Market impacted?5. What is the competitive landscape, who are the key players?6. What are some of the recent M&A, PE / VC deals that have happened in the Circulating Tumor Cells (CTC) Market?

The report also analysis the impact of COVID 19 based on a scenario-based modelling. This provides a clear view of how has COVID impacted the growth cycle & when is the likely recovery of the industry is expected to pre-covid levels.

Contact us:i2iResearch info to intelligenceLocational Office: *India, *United State, *GermanyEmail: [emailprotected]Toll-free: +1-800-419-8865 | Phone: +91 98801 53667

Read the original:
Circulating Tumor Cells (CTC) Market Growth Analysis and Forecast to 2026 by Applications and Competitors - Cheshire Media

Read More...

UPDATED: Merck’s Keytruda nets another approval, this time in triple negative breast cancer. Can it catch up to Tecentriq? – Endpoints News

Tuesday, November 17th, 2020

Another day, another win for Mercks blockbuster Keytruda.

The FDA has granted accelerated approval for the cash cow combined with chemotherapy in triple negative breast cancer, giving the drug the green light in its 18th different cancer. Mondays new indication comes for patients with PD-L1-expressing tumors with a Combined Positive Score of at least 10.

Merck noted that due to the nature of the accelerated approval, the thumbs up is contingent upon confirmatory trials.

Data for the approval first came back in February, when the Keynote-355 trial demonstrated Keytruda plus chemo significantly improved progression-free survival compared to chemo by itself. The study showed that, in the target population with a CPS of at least 10, the combination reduced the risk of disease progression or death by 35% with a median PFS of 9.7 months, against 5.6 months in the placebo arm.

On safety, the February data showed 2.5% of all patients in the drug arm saw fatal adverse events, including cardiac arrest and septic shock, with serious side effects appearing in 30% of patients. Keytruda was discontinued due to adverse events in 11% of patients.

Frontline triple negative breast cancer is a particularly difficult indication to treat, as the growth of the cancer is not fueled by the hormones estrogen and progesterone, or by the HER2 protein. Its one of the rare fields in which Roches PD-L1 Tecentriq has enjoyed a head start over Keytruda and Opdivo, the leaders in the checkpoint race, as Tecentriq is approved in combination with Abraxane for this indication.

Back in May 2019, Merck conceded a failure in the arena after a Phase III study flopped on overall survival. But a few months later, the pharma turned things around after discovering a neoadjuvant regimen of Keytruda and chemo followed by Keytruda monotherapy after surgery induced a higher pathological complete response rate.

Though execs presented that as a positive, some analysts didnt paint as sunny a picture. This past February, when the Keynote-355 topline data was first published, SVB Leerinks Daina Graybosch pointed out that because only patients with a CPS of at least 10 appeared to benefit, instead of a score of at least 1, it wont be able to treat as broad a population as Tecentriq. Roche, she noted, also has about a two-year head start.

A Merck spokesperson also had this to say about the CPS and IC percentages:

In TNBC, we measure PD-L1 with a combined positive score (CPS). The CPS includes staining for tumor cells, as well as tumor-infiltrating immune cells and it is not a percentage. We believe CPS 10 is roughly equivalent to how Roche scores PD-L1+ patients (IC>=1% based on the SP142 assay) on tumor-infiltrating immune cells (IC). The prevalence of the PD-L1 positive population in TNBC whether by CPS of greater than or equal to 10 or IC of 1% is both about 40%.

Keytruda is already one of the best-selling drugs in the world, having notched roughly $3.9 billion in the first half of 2020 alone. Some have predicted the drug may overtake AbbVies Humira as the top seller within the next few years, with the most optimistic estimate pegged for $22.2 billion in sales by 2025.

Here is the original post:
UPDATED: Merck's Keytruda nets another approval, this time in triple negative breast cancer. Can it catch up to Tecentriq? - Endpoints News

Read More...

The CHMP has adopted a negative opinion for emapalumab in Europe for the treatment of primary HLH | Antibodies | News Channels – PipelineReview.com

Tuesday, November 17th, 2020

DetailsCategory: AntibodiesPublished on Friday, 13 November 2020 13:04Hits: 719

STOCKHOLM, Sweden I November 13, 2020 I Swedish Orphan Biovitrum AB (publ) (Sobi) (STO:SOBI) today announced that the Committee for Medicinal Products for Human use (CHMP) has adopted a negative opinion recommending a refusal of the marketing authorisation for emapalumab for the treatment of primary haemophagocytic lymphohistiocytosis (pHLH) in children under 18 years of age in Europe. The negative opinion was given after the re-examination requested by Sobi after the initial opinon in July 2020.

This recommendation by the CHMP is disappointing given the significant unmet medical need which exists for patients with pHLH who have no approved therapies in Europe. During the re-examination we worked extensively with physicians and patients and were able to resolve some but not all of the concerns raised by EMA, said Ravi Rao, Head of R&D and Chief Medical Officer at Sobi. We are confident about the clinical profile of emapalumab and our focus is now on increasing access for patients in other regions and developing new indications for this medicine.

About primary HLHPrimary HLH is a rare syndrome that typically presents in infancy but can also be seen in adults and is associated with high morbidity and mortality. In spite of some treatment advances, there continues to be a very high unmet medical need in particular in patients that have failed conventional therapy as there are no approved treatment options outside the US. In the US, emapalumab is the first therapy approved by the US Food & Drug Administration (FDA) for primary HLH. Over 100 patients have been treated in the US and the benefit/risk profile continues to be favourable.

About emapalumabEmapalumab is a monoclonal antibody that binds to and neutralises interferon gamma (IFN). In the US, emapalumab is indicated for the treatment of adult and paediatric (new-born and older) patients with primary hemophagocytic lymphohistiocytosis (HLH) with refractory, recurrent or progressive disease or intolerance with conventional HLH therapy. Primary HLH is a rare syndrome of hyperinflammation that usually occurs within the first year of life and can rapidly become fatal unless diagnosed and treated. The FDA approval is based on data from the phase 2/3 studies (NCT01818492 and NCT02069899). Emapalumab is indicated for administration through intravenous infusion over one hour twice per week until haematopoietic stem cell transplantation (HSCT). For more information please see http://www.gamifant.com including the full US Prescribing Information. In September 2020, emapalumab received Orphan Drug Designation (ODD) by the FDA for prevention of graft failure following haematopoietic stem cell transplantation.

About SobiTMSobi is a specialised international biopharmaceutical company transforming the lives of people with rare diseases. Sobi is providing sustainable access to innovative therapies in the areas of haematology, immunology and specialty indications. Today, Sobi employs approximately 1,400 people across Europe, North America, the Middle East, Russia and North Africa. In 2019, Sobis revenues amounted to SEK 14.2 billion. Sobis share (STO:SOBI) is listed on Nasdaq Stockholm. You can find more information about Sobi at http://www.sobi.com.

SOURCE: Sobi

Continue reading here:
The CHMP has adopted a negative opinion for emapalumab in Europe for the treatment of primary HLH | Antibodies | News Channels - PipelineReview.com

Read More...

Taking Cultured Meat to the Next Level – Technology Networks

Tuesday, November 17th, 2020

With its origins in the late 1990s, lab-grown or cultured meat, is produced by providing stem cells extracted from the muscle of an animal with a suitable growth medium and nutrients, enabling them to proliferate and then differentiate to form muscle tissue. Creating meat in this way could help to address some of the environmental and ethical issues associated with livestock farming, as well as offer health benefits to consumers.Pairing cellular agriculture with genetic engineering could also enable the development of novel foods, with non-native features, that may be nutritionally enhanced. In a study recently published in Metabolic Engineering, researchers from Tufts University engineered bovine cells to endogenously produce phytoene, lycopene and -carotene, and found a reduction in lipid oxidation levels when this cultured meat was cooked.

Technology Networks had the pleasure of speaking to Andrew Stout, lead author of the study, to learn how these cells were created and explore the benefits of engineering in the abilities to produce additional nutrients. Andrew also discussed some of the challenges that are so far limiting the wider commercialization of cultured meat and how this may change in the future.

Anna MacDonald (AM): Can you give us an overview of the process by which the cow cells were engineered to produce the carotenoids?Andrew Stout (AS): To do this, we inserted three genes into the cells which encode enzymes that convert native compounds into carotenoids. Specifically, the first gene in this pathway (phytoene synthase) takes a native chemical and turns it into the carotenoid phytoene. The second gene (phytoene desaturase) turns some of that phytoene into a second carotenoid called lycopene. And the third gene (lycopene cyclase) turns some of that lycopene into a third carotenoid called beta-carotene. In that way, we're able to get cow cells to produce three different carotenoids that aren't naturally produced in bovine tissue. To engineer the cells, we used a system called the Sleeping beauty transposon system. This system is essentially a "cut and paste" tool which randomly cuts open the cells' genomes and inserts new DNA which we provide (in this case, the genes for carotenoid-producing enzymes).

AM: Why were beta-carotene, phytoene and lycopene chosen in particular?AS: There were several reasons for this. The first and most important was their role as dietary antioxidants. A key mechanistic link between red meat consumption and colorectal cancer is through lipid oxidation. This oxidation leads to the production of free radicals that can interact with tissue in the colon, damage cellular DNA, and ultimately contribute to cancer formation. Antioxidants can act to "quench" those free radicals, thus potentially inhibiting their cancer-causing potential. As carotenoids are powerful antioxidants, they offer a promising target for improving the nutritional features of cell-cultured meat.

Other reasons include the importance of beta-carotene as a vitamin A precursor, previous demonstrations of phytoene synthase efficacy in mammalian cells, and also as a sort of homage to golden rice, the first major demonstration of using genetic engineering to nutritionally enhance a food product.

AM: Were there any side-effects as a result of the nutritional engineering?AS: There were a few. The most obvious was a reduction in growth rate in bovine satellite cells that were engineered with carotenoid-producing enzymes. This would have negative implications for production processes if it proves to be unavoidable. Interestingly, though, in immortalized mouse muscle cells, this reduction in growth rate wasn't seen. Instead, cells producing carotenoids actually grew faster than non-engineered cells. One explanation for this could be that immortalized cells are more "robust" and are more amenable to engineering than the primary (non-immortalized) cow muscle cells we used. It's possible that immortalized cow cells would show growth-effects more like those seen in the mouse cells, which would turn this production down-side into a production up-side. Another side effect we saw was a change in color of the cells -- they took on a reddish tinge with the production of the carotenoids. I don't think this is really a "positive" or "negative" effect, but it is pretty interesting. Other potential side-effects that would need to be looked into would be the effects of carotenoids on cell differentiation, on the prevalence of other nutrients (e.g., cholesterol, etc.), and on flavor, texture, aroma, etc.

Karen Steward (KS): Why do you think you saw lower levels of lipid oxidation when the cell cultured meat was cooked compared to conventional meat?AS: Since carotenoids are antioxidants, they act to quench oxidation in cells during storage, cooking, etc., so we would expect lower lipid oxidation if the cells are producing carotenoids and therefore increasing the total cellular level of antioxidants.

KS: What do you see are the benefits of engineering in the abilities to produce additional phytonutrients to beef cells, as opposed say to having a traditional steak with some vegetables? Is there a risk that in providing these nutrients through meat intake a diet would consequently lack fibre which could impact gut health?AS: This is a fun question! I think we're an extremely long way from actually being able to use this technology to replace vegetables on our plates (and anyways, what a culinarily boring world that would be!) I like to think of this technology not as a replacement of vegetables, but an enhancement of meat. For instance, not all vegetables are high in carotenoids, so if you can get those nutrients from another source in your meal, then your overall consumption of them can increase. Also, the roll of carotenoids in specifically inhibiting oxidation in meat can act to mitigate some of the negative health implications of meat consumption without aiming to reduce vegetable consumption. As a final note, I'd like to think of this work as really just the tip of the iceberg of what's possible. There are so many options for enhancing meat with this or similar technologies--enhanced flavor, therapeutic activity, enhanced smell, etc. I think there's a world of totally novel foods that are possible and that would expand our culinary palette, not reduce it.

KS: Is there any need to start with cow cells? Could you essentially start with any cell type or are there limitations?AS: No need at all! I think this would likely work for all mammalian cells, and there's a strong chance it would work for avian and fish cells as well. We wanted to work with bovine cells because beef is such a major contributor to meat-associated greenhouse gas production and is one of the main red meats consumed around the world. As such, I think it's a really important target for all cultured meat work, including nutritional engineering.

AM: What challenges are so far limiting the wider commercialization of cultured meat?AS: The key hurdles are cost and scale. The field needs to reduce the cost of growth media (likely by reducing the cost of growth factors, reducing cellular reliance on growth factors, finding growth-factor alternatives, or other creative solutions), and to increase the scalability of cell culture (increased growth rate, increased maximum cell density, etc.). There are certainly plenty of other challenges, such as regulatory and consumer reception, demonstration of nutritional and food-quality value, and demonstration of food-safety, but I think that right now cost and scale still reign supreme.

AM: Where do you see the future of cellular agriculture headed?AS: A good question! I'll answer for two slightly different technologies.

First, for cultured meat specifically:

I think in the near future, the field is heading towards a bit of a "realignment" or specification in terms of goals, expectations, hype, etc. I think that this can be seen in some of the ways that companies are starting to look at their products with a bit more nuance, such as looking at the possibility of hybrid cell-based/plant-based products, which could overcome some of the cost/scale barriers of a fully cell-cultured product. Beyond that, I like to think that there will be an expansion of creative solutions to problems, or creative new ways of thinking about cell cultured meat. This could come in the form of looking at agricultural waste products for cell culture components, exploring novel genetic strategies to improve growth / reduce cost, or looking into alternate culture strategies / bioreactors.

Then for cellular agriculture more generally:

I think cellular agriculture in general, while certainly offering its own challenges and hurdles, is a lot further along the developmental pathway than cultured meat. I'm thinking here of products that are already on the market and demonstrably feasible such as recombinant milk proteins (Perfect Day Foods), recombinant collagen proteins (Geltor, Inc.), or recombinant proteins to improve plant-based products (Impossible Foods). I think these technologies are going to continue coming out and coming down in cost, allowing a bunch of new awesome products to come out and accelerate the development of plant-based or fermentation-derived products.

Andrew Stout was speaking to Anna MacDonald and Dr Karen Steward, Science Writers for Technology Networks.

Read the original here:
Taking Cultured Meat to the Next Level - Technology Networks

Read More...

FDA Approves Merck’s KEYTRUDA (pembrolizumab) in Combination With Chemotherapy for Patients With Locally Recurrent Unresectable or Metastatic…

Tuesday, November 17th, 2020

Approximately 15-20% of patients with breast cancer are diagnosed with triple-negative breast cancer, which is a difficult-to-treat and aggressive cancer, said Dr. Hope Rugo, director of Breast Oncology and Clinical Trials Education, University of California San Francisco (UCSF) Helen Diller Family Comprehensive Cancer Center. Notably, in KEYNOTE-355, KEYTRUDA was combined with three different chemotherapy regimens: paclitaxel, nab-paclitaxel or gemcitabine and carboplatin. The approval of KEYTRUDA in combination with chemotherapy gives physicians an important new option for appropriate patients.

Immune-mediated adverse reactions, which may be severe or fatal, can occur with KEYTRUDA, including pneumonitis, colitis, hepatitis, endocrinopathies, nephritis, severe skin reactions, solid organ transplant rejection, and complications of allogeneic hematopoietic stem cell transplantation. Based on the severity of the adverse reaction, KEYTRUDA should be withheld or discontinued and corticosteroids administered if appropriate. KEYTRUDA can also cause severe or life-threatening infusion-related reactions. Based on its mechanism of action, KEYTRUDA can cause fetal harm when administered to a pregnant woman. For more information, see Selected Important Safety Information below.

Todays approval is a significant milestone, as it represents the first approval for KEYTRUDA in the breast cancer setting, said Dr. Roy Baynes, senior vice president and head of global clinical development, chief medical officer, Merck Research Laboratories. In the study supporting this approval, KEYTRUDA in combination with paclitaxel, nab-paclitaxel or gemcitabine and carboplatin significantly improved progression-free survival for patients with advanced triple-negative breast cancer whose tumors express PD-L1 with CPS greater than or equal to 10 compared with the same chemotherapy regimens alone.

Data Supporting the Approval

The accelerated approval was based on data from KEYNOTE-355 (ClinicalTrials.gov, NCT02819518), a multicenter, double-blind, randomized, placebo-controlled trial conducted in 847 patients with locally recurrent unresectable or metastatic TNBC, regardless of tumor PD-L1 expression, who had not been previously treated with chemotherapy in the metastatic setting. Patients were randomized (2:1) to receive either KEYTRUDA (200 mg on Day 1 every three weeks) or placebo (on Day 1 every three weeks) in combination with the following chemotherapy; all medications were administered via intravenous infusion:

Randomization was stratified by chemotherapy treatment (pac or nab-paclitaxel vs. gem and carbo), tumor PD-L1 expression (CPS 1 vs. CPS <1) according to the PD-L1 IHC 22C3 pharmDx kit and prior treatment with the same class of chemotherapy in the neoadjuvant setting (yes vs. no). Assessment of tumor status was performed at Weeks 8, 16 and 24, then every nine weeks for the first year and every 12 weeks thereafter. The main efficacy outcome measure was PFS as assessed by blinded independent central review (BICR) according to RECIST v1.1, modified to follow a maximum of 10 target lesions and a maximum of five target lesions per organ tested in the subgroup of patients with CPS 10. Additional efficacy outcome measures were overall survival, as well as objective response rate (ORR) and duration of response (DOR) as assessed by BICR.

The study population characteristics were: median age of 53 years (range, 22 to 85), 21% age 65 or older; 100% female; 68% White, 21% Asian and 4% Black; 60% ECOG PS of 0 and 40% ECOG PS of 1; and 68% were post-menopausal. Seventy-five percent of patients had tumor PD-L1 expression CPS 1 and 38% had tumor PDL1 expression CPS 10.

In KEYNOTE-355, efficacy results were in patients who were PDL1 positive with a CPS 10 (n=323) and randomized to receive KEYTRUDA in combination with pac, nab-paclitaxel or gem/carbo compared with the same chemotherapy regimens alone. KEYTRUDA in combination with pac, nab-paclitaxel or gem/carbo (n=220) reduced the risk of disease progression or death by 35% (HR=0.65 [95% CI, 0.49, 0.86]; p=0.0012), with a median PFS of 9.7 months (95% CI, 7.6, 11.3) versus 5.6 months (95% CI, 5.3, 7.5) with the same chemotherapy regimens alone (n=103). For PFS, 62% (n=136) of patients experienced an event with KEYTRUDA in combination with pac, nab-paclitaxel or gem/carbo versus 77% (n=79) with the same chemotherapy regimens alone. For patients who received KEYTRUDA in combination with pac, nab-paclitaxel or gem/carbo, the ORR was 53% (95% CI, 46, 60), with a complete response rate of 17% and a partial response rate of 36%. For patients treated with the same chemotherapy regimens alone, the ORR was 40% (95% CI, 30, 50), with a complete response rate of 13% and a partial response rate of 27%. Median DOR was 19.3 months (95% CI, 9.9, 29.8) with KEYTRUDA in combination with pac, nab-paclitaxel or gem/carbo versus 7.3 months (95% CI, 5.3, 15.8) with the same chemotherapy regimens alone.

In the study, the median duration of exposure to KEYTRUDA was 5.7 months (range, 1 day to 33.0 months). Fatal adverse reactions occurred in 2.5% of patients (n=596) receiving KEYTRUDA in combination with chemotherapy, including cardio-respiratory arrest (0.7%) and septic shock (0.3%). Serious adverse reactions occurred in 30% of patients receiving KEYTRUDA in combination with pac, nab-paclitaxel, or gem/carbo. Serious adverse reactions observed in 2% of patients were pneumonia (2.9%), anemia (2.2%), and thrombocytopenia (2%). KEYTRUDA was discontinued for adverse reactions in 11% of patients. The most common adverse reactions resulting in permanent discontinuation of KEYTRUDA (1%) were increased alanine aminotransferase (ALT) (2.2%), increased aspartate aminotransferase (AST) (1.5%), and pneumonitis (1.2%). Adverse reactions leading to the interruption of KEYTRUDA occurred in 50% of patients. The most common adverse reactions leading to interruption of KEYTRUDA (2%) were neutropenia (22%), thrombocytopenia (14%), anemia (7%), increased ALT (6%), leukopenia (5%), decreased white blood cell count (3.9%), and diarrhea (2%). The most common adverse reactions (all grades 20%) for KEYTRUDA in combination with pac, nab-paclitaxel or gem/carbo were: fatigue (48%), nausea (44%), alopecia (34%), diarrhea and constipation (28% each), vomiting and rash (26% each), cough (23%), decreased appetite (21%), and headache (20%).

About Triple-Negative Breast Cancer (TNBC)

Triple-negative breast cancer is an aggressive type of breast cancer that characteristically has a high recurrence rate within the first five years after diagnosis. While some breast cancers may test positive for estrogen receptors, progesterone receptors or overexpression of human epidermal growth factor receptor 2 (HER2), TNBC tests negative for all three. Approximately 15-20% of patients with breast cancer are diagnosed with TNBC.

About KEYTRUDA (pembrolizumab) Injection, 100 mg

KEYTRUDA is an anti-PD-1 therapy that works by increasing the ability of the bodys immune system to help detect and fight tumor cells. KEYTRUDA is a humanized monoclonal antibody that blocks the interaction between PD-1 and its ligands, PD-L1 and PD-L2, thereby activating T lymphocytes which may affect both tumor cells and healthy cells.

Merck has the industrys largest immuno-oncology clinical research program. There are currently more than 1,300 trials studying KEYTRUDA across a wide variety of cancers and treatment settings. The KEYTRUDA clinical program seeks to understand the role of KEYTRUDA across cancers and the factors that may predict a patient's likelihood of benefitting from treatment with KEYTRUDA, including exploring several different biomarkers.

Selected KEYTRUDA (pembrolizumab) Indications in the U.S.

Melanoma

KEYTRUDA is indicated for the treatment of patients with unresectable or metastatic melanoma.

KEYTRUDA is indicated for the adjuvant treatment of patients with melanoma with involvement of lymph node(s) following complete resection.

Non-Small Cell Lung Cancer

KEYTRUDA, in combination with pemetrexed and platinum chemotherapy, is indicated for the first-line treatment of patients with metastatic nonsquamous non-small cell lung cancer (NSCLC), with no EGFR or ALK genomic tumor aberrations.

KEYTRUDA, in combination with carboplatin and either paclitaxel or paclitaxel protein-bound, is indicated for the first-line treatment of patients with metastatic squamous NSCLC.

KEYTRUDA, as a single agent, is indicated for the first-line treatment of patients with NSCLC expressing PD-L1 [tumor proportion score (TPS) 1%] as determined by an FDA-approved test, with no EGFR or ALK genomic tumor aberrations, and is stage III where patients are not candidates for surgical resection or definitive chemoradiation, or metastatic.

KEYTRUDA, as a single agent, is indicated for the treatment of patients with metastatic NSCLC whose tumors express PD-L1 (TPS 1%) as determined by an FDA-approved test, with disease progression on or after platinum-containing chemotherapy. Patients with EGFR or ALK genomic tumor aberrations should have disease progression on FDA-approved therapy for these aberrations prior to receiving KEYTRUDA.

Small Cell Lung Cancer

KEYTRUDA is indicated for the treatment of patients with metastatic small cell lung cancer (SCLC) with disease progression on or after platinum-based chemotherapy and at least 1 other prior line of therapy. This indication is approved under accelerated approval based on tumor response rate and durability of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in confirmatory trials.

Head and Neck Squamous Cell Cancer

KEYTRUDA, in combination with platinum and fluorouracil (FU), is indicated for the first-line treatment of patients with metastatic or with unresectable, recurrent head and neck squamous cell carcinoma (HNSCC).

KEYTRUDA, as a single agent, is indicated for the first-line treatment of patients with metastatic or with unresectable, recurrent HNSCC whose tumors express PD-L1 [combined positive score (CPS) 1] as determined by an FDA-approved test.

KEYTRUDA, as a single agent, is indicated for the treatment of patients with recurrent or metastatic HNSCC with disease progression on or after platinum-containing chemotherapy.

Classical Hodgkin Lymphoma

KEYTRUDA is indicated for the treatment of adult patients with relapsed or refractory classical Hodgkin lymphoma (cHL).

KEYTRUDA is indicated for the treatment of pediatric patients with refractory cHL, or cHL that has relapsed after 2 or more lines of therapy.

Primary Mediastinal Large B-Cell Lymphoma

KEYTRUDA is indicated for the treatment of adult and pediatric patients with refractory primary mediastinal large B-cell lymphoma (PMBCL), or who have relapsed after 2 or more prior lines of therapy. KEYTRUDA is not recommended for treatment of patients with PMBCL who require urgent cytoreductive therapy.

Urothelial Carcinoma

KEYTRUDA is indicated for the treatment of patients with locally advanced or metastatic urothelial carcinoma (mUC) who are not eligible for cisplatin-containing chemotherapy and whose tumors express PD-L1 (CPS 10), as determined by an FDA-approved test, or in patients who are not eligible for any platinum-containing chemotherapy regardless of PD-L1 status. This indication is approved under accelerated approval based on tumor response rate and duration of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in confirmatory trials.

KEYTRUDA is indicated for the treatment of patients with locally advanced or metastatic urothelial carcinoma (mUC) who have disease progression during or following platinum-containing chemotherapy or within 12 months of neoadjuvant or adjuvant treatment with platinum-containing chemotherapy.

KEYTRUDA is indicated for the treatment of patients with Bacillus Calmette-Guerin (BCG)-unresponsive, high-risk, non-muscle invasive bladder cancer (NMIBC) with carcinoma in situ (CIS) with or without papillary tumors who are ineligible for or have elected not to undergo cystectomy.

Microsatellite Instability-High or Mismatch Repair Deficient Cancer

KEYTRUDA is indicated for the treatment of adult and pediatric patients with unresectable or metastatic microsatellite instability-high (MSI-H) or mismatch repair deficient (dMMR)

This indication is approved under accelerated approval based on tumor response rate and durability of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials. The safety and effectiveness of KEYTRUDA in pediatric patients with MSI-H central nervous system cancers have not been established.

Microsatellite Instability-High or Mismatch Repair Deficient Colorectal Cancer

KEYTRUDA is indicated for the first-line treatment of patients with unresectable or metastatic MSI-H or dMMR colorectal cancer (CRC).

Gastric Cancer

KEYTRUDA is indicated for the treatment of patients with recurrent locally advanced or metastatic gastric or gastroesophageal junction (GEJ) adenocarcinoma whose tumors express PD-L1 (CPS 1) as determined by an FDA-approved test, with disease progression on or after two or more prior lines of therapy including fluoropyrimidine- and platinum-containing chemotherapy and if appropriate, HER2/neu-targeted therapy. This indication is approved under accelerated approval based on tumor response rate and durability of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials.

Esophageal Cancer

KEYTRUDA is indicated for the treatment of patients with recurrent locally advanced or metastatic squamous cell carcinoma of the esophagus whose tumors express PD-L1 (CPS 10) as determined by an FDA-approved test, with disease progression after one or more prior lines of systemic therapy.

Cervical Cancer

KEYTRUDA is indicated for the treatment of patients with recurrent or metastatic cervical cancer with disease progression on or after chemotherapy whose tumors express PD-L1 (CPS 1) as determined by an FDA-approved test. This indication is approved under accelerated approval based on tumor response rate and durability of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials.

Hepatocellular Carcinoma

KEYTRUDA is indicated for the treatment of patients with hepatocellular carcinoma (HCC) who have been previously treated with sorafenib. This indication is approved under accelerated approval based on tumor response rate and durability of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials.

Merkel Cell Carcinoma

KEYTRUDA is indicated for the treatment of adult and pediatric patients with recurrent locally advanced or metastatic Merkel cell carcinoma (MCC). This indication is approved under accelerated approval based on tumor response rate and durability of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials.

Renal Cell Carcinoma

KEYTRUDA, in combination with axitinib, is indicated for the first-line treatment of patients with advanced renal cell carcinoma (RCC).

Tumor Mutational Burden-High

KEYTRUDA is indicated for the treatment of adult and pediatric patients with unresectable or metastatic tumor mutational burden-high (TMB-H) [10 mutations/megabase] solid tumors, as determined by an FDA-approved test, that have progressed following prior treatment and who have no satisfactory alternative treatment options. This indication is approved under accelerated approval based on tumor response rate and durability of response. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials. The safety and effectiveness of KEYTRUDA in pediatric patients with TMB-H central nervous system cancers have not been established.

Cutaneous Squamous Cell Carcinoma

KEYTRUDA is indicated for the treatment of patients with recurrent or metastatic cutaneous squamous cell carcinoma (cSCC) that is not curable by surgery or radiation.

Triple-Negative Breast Cancer

KEYTRUDA, in combination with chemotherapy, is indicated for the treatment of patients with locally recurrent unresectable or metastatic triple-negative breast cancer (TNBC) whose tumors express PD-L1 (CPS 10) as determined by an FDA-approved test. This indication is approved under accelerated approval based on progression-free survival. Continued approval for this indication may be contingent upon verification and description of clinical benefit in the confirmatory trials.

Selected Important Safety Information for KEYTRUDA

Severe and Fatal Immune-Mediated Adverse Reactions

KEYTRUDA is a monoclonal antibody that belongs to a class of drugs that bind to either the programmed death receptor-1 (PD-1) or the programmed death ligand 1 (PD-L1), blocking the PD-1/PD-L1 pathway, thereby removing inhibition of the immune response, potentially breaking peripheral tolerance and inducing immune-mediated adverse reactions. Immune-mediated adverse reactions, which may be severe or fatal, can occur in any organ system or tissue, can affect more than one body system simultaneously, and can occur at any time after starting treatment or after discontinuation of treatment.

Monitor patients closely for symptoms and signs that may be clinical manifestations of underlying immune-mediated adverse reactions. Early identification and management are essential to ensure safe use of antiPD-1/PD-L1 treatments. Evaluate liver enzymes, creatinine, and thyroid function at baseline and periodically during treatment. In cases of suspected immune-mediated adverse reactions, initiate appropriate workup to exclude alternative etiologies, including infection. Institute medical management promptly, including specialty consultation as appropriate.

Withhold or permanently discontinue KEYTRUDA depending on severity of the immune-mediated adverse reaction. In general, if KEYTRUDA requires interruption or discontinuation, administer systemic corticosteroid therapy (1 to 2 mg/kg/day prednisone or equivalent) until improvement to Grade 1 or less. Upon improvement to Grade 1 or less, initiate corticosteroid taper and continue to taper over at least 1 month. Consider administration of other systemic immunosuppressants in patients whose adverse reactions are not controlled with corticosteroid therapy.

Immune-Mediated Pneumonitis

KEYTRUDA can cause immune-mediated pneumonitis. The incidence is higher in patients who have received prior thoracic radiation. Immune-mediated pneumonitis occurred in 3.4% (94/2799) of patients receiving KEYTRUDA, including fatal (0.1%), Grade 4 (0.3%), Grade 3 (0.9%), and Grade 2 (1.3%) reactions. Systemic corticosteroids were required in 67% (63/94) of patients. Pneumonitis led to permanent discontinuation of KEYTRUDA in 1.3% (36) and withholding in 0.9% (26) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement; of these, 23% had recurrence. Pneumonitis resolved in 59% of the 94 patients.

Pneumonitis occurred in 8% (31/389) of adult patients with cHL receiving KEYTRUDA as a single agent, including Grades 3-4 in 2.3% of patients. Patients received high-dose corticosteroids for a median duration of 10 days (range: 2 days to 53 months). Pneumonitis rates were similar in patients with and without prior thoracic radiation. Pneumonitis led to discontinuation of KEYTRUDA in 5.4% (21) of patients, 42% of these patients interrupted KEYTRUDA, 68% discontinued KEYTRUDA, and 77% had resolution.

Immune-Mediated Colitis

KEYTRUDA can cause immune-mediated colitis, which may present with diarrhea. Cytomegalovirus infection/reactivation has been reported in patients with corticosteroid-refractory immune-mediated colitis. In cases of corticosteroid-refractory colitis, consider repeating infectious workup to exclude alternative etiologies. Immune-mediated colitis occurred in 1.7% (48/2799) of patients receiving KEYTRUDA, including Grade 4 (<0.1%), Grade 3 (1.1%), and Grade 2 (0.4%) reactions. Systemic corticosteroids were required in 69% (33/48); additional immunosuppressant therapy was required in 4.2% of patients. Colitis led to permanent discontinuation of KEYTRUDA in 0.5% (15) and withholding in 0.5% (13) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement; of these, 23% had recurrence. Colitis resolved in 85% of the 48 patients.

Hepatotoxicity and Immune-Mediated Hepatitis

KEYTRUDA as a Single Agent

KEYTRUDA can cause immune-mediated hepatitis. Immune-mediated hepatitis occurred in 0.7% (19/2799) of patients receiving KEYTRUDA, including Grade 4 (<0.1%), Grade 3 (0.4%), and Grade 2 (0.1%) reactions. Systemic corticosteroids were required in 68% (13/19) of patients; additional immunosuppressant therapy was required in 11% of patients. Hepatitis led to permanent discontinuation of KEYTRUDA in 0.2% (6) and withholding in 0.3% (9) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement; of these, none had recurrence. Hepatitis resolved in 79% of the 19 patients.

KEYTRUDA with Axitinib

KEYTRUDA in combination with axitinib can cause hepatic toxicity. Monitor liver enzymes before initiation of and periodically throughout treatment. Consider monitoring more frequently as compared to when the drugs are administered as single agents. For elevated liver enzymes, interrupt KEYTRUDA and axitinib, and consider administering corticosteroids as needed. With the combination of KEYTRUDA and axitinib, Grades 3 and 4 increased alanine aminotransferase (ALT) (20%) and increased aspartate aminotransferase (AST) (13%) were seen, which was at a higher frequency compared to KEYTRUDA alone. Fifty-nine percent of the patients with increased ALT received systemic corticosteroids. In patients with ALT 3 times upper limit of normal (ULN) (Grades 2-4, n=116), ALT resolved to Grades 0-1 in 94%. Among the 92 patients who were rechallenged with either KEYTRUDA (n=3) or axitinib (n=34) administered as a single agent or with both (n=55), recurrence of ALT 3 times ULN was observed in 1 patient receiving KEYTRUDA, 16 patients receiving axitinib, and 24 patients receiving both. All patients with a recurrence of ALT 3 ULN subsequently recovered from the event.

Immune-Mediated Endocrinopathies

Adrenal Insufficiency

KEYTRUDA can cause primary or secondary adrenal insufficiency. For Grade 2 or higher, initiate symptomatic treatment, including hormone replacement as clinically indicated. Withhold KEYTRUDA depending on severity. Adrenal insufficiency occurred in 0.8% (22/2799) of patients receiving KEYTRUDA, including Grade 4 (<0.1%), Grade 3 (0.3%), and Grade 2 (0.3%) reactions. Systemic corticosteroids were required in 77% (17/22) of patients; of these, the majority remained on systemic corticosteroids. Adrenal insufficiency led to permanent discontinuation of KEYTRUDA in <0.1% (1) and withholding in 0.3% (8) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement.

Hypophysitis

KEYTRUDA can cause immune-mediated hypophysitis. Hypophysitis can present with acute symptoms associated with mass effect such as headache, photophobia, or visual field defects. Hypophysitis can cause hypopituitarism. Initiate hormone replacement as indicated. Withhold or permanently discontinue KEYTRUDA depending on severity. Hypophysitis occurred in 0.6% (17/2799) of patients receiving KEYTRUDA, including Grade 4 (<0.1%), Grade 3 (0.3%), and Grade 2 (0.2%) reactions. Systemic corticosteroids were required in 94% (16/17) of patients; of these, the majority remained on systemic corticosteroids. Hypophysitis led to permanent discontinuation of KEYTRUDA in 0.1% (4) and withholding in 0.3% (7) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement.

Thyroid Disorders

KEYTRUDA can cause immune-mediated thyroid disorders. Thyroiditis can present with or without endocrinopathy. Hypothyroidism can follow hyperthyroidism. Initiate hormone replacement for hypothyroidism or institute medical management of hyperthyroidism as clinically indicated. Withhold or permanently discontinue KEYTRUDA depending on severity. Thyroiditis occurred in 0.6% (16/2799) of patients receiving KEYTRUDA, including Grade 2 (0.3%). None discontinued, but KEYTRUDA was withheld in <0.1% (1) of patients.

Hyperthyroidism occurred in 3.4% (96/2799) of patients receiving KEYTRUDA, including Grade 3 (0.1%) and Grade 2 (0.8%). It led to permanent discontinuation of KEYTRUDA in <0.1% (2) and withholding in 0.3% (7) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement. Hypothyroidism occurred in 8% (237/2799) of patients receiving KEYTRUDA, including Grade 3 (0.1%) and Grade 2 (6.2%). It led to permanent discontinuation of KEYTRUDA in <0.1% (1) and withholding in 0.5% (14) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement. The majority of patients with hypothyroidism required long-term thyroid hormone replacement. The incidence of new or worsening hypothyroidism was higher in 1185 patients with HNSCC, occurring in 16% of patients receiving KEYTRUDA as a single agent or in combination with platinum and FU, including Grade 3 (0.3%) hypothyroidism. The incidence of new or worsening hypothyroidism was higher in 389 adult patients with cHL (17%) receiving KEYTRUDA as a single agent, including Grade 1 (6.2%) and Grade 2 (10.8%) hypothyroidism.

Type 1 Diabetes Mellitus (DM), Which Can Present With Diabetic Ketoacidosis

Monitor patients for hyperglycemia or other signs and symptoms of diabetes. Initiate treatment with insulin as clinically indicated. Withhold KEYTRUDA depending on severity. Type 1 DM occurred in 0.2% (6/2799) of patients receiving KEYTRUDA. It led to permanent discontinuation in <0.1% (1) and withholding of KEYTRUDA in <0.1% (1). All patients who were withheld reinitiated KEYTRUDA after symptom improvement.

Immune-Mediated Nephritis With Renal Dysfunction

KEYTRUDA can cause immune-mediated nephritis. Immune-mediated nephritis occurred in 0.3% (9/2799) of patients receiving KEYTRUDA, including Grade 4 (<0.1%), Grade 3 (0.1%), and Grade 2 (0.1%) reactions. Systemic corticosteroids were required in 89% (8/9) of patients. Nephritis led to permanent discontinuation of KEYTRUDA in 0.1% (3) and withholding in 0.1% (3) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement; of these, none had recurrence. Nephritis resolved in 56% of the 9 patients.

Immune-Mediated Dermatologic Adverse Reactions

KEYTRUDA can cause immune-mediated rash or dermatitis. Exfoliative dermatitis, including Stevens-Johnson syndrome, drug rash with eosinophilia and systemic symptoms, and toxic epidermal necrolysis, has occurred with antiPD-1/PD-L1 treatments. Topical emollients and/or topical corticosteroids may be adequate to treat mild to moderate nonexfoliative rashes. Withhold or permanently discontinue KEYTRUDA depending on severity. Immune-mediated dermatologic adverse reactions occurred in 1.4% (38/2799) of patients receiving KEYTRUDA, including Grade 3 (1%) and Grade 2 (0.1%) reactions. Systemic corticosteroids were required in 40% (15/38) of patients. These reactions led to permanent discontinuation in 0.1% (2) and withholding of KEYTRUDA in 0.6% (16) of patients. All patients who were withheld reinitiated KEYTRUDA after symptom improvement; of these, 6% had recurrence. The reactions resolved in 79% of the 38 patients.

Other Immune-Mediated Adverse Reactions

The following clinically significant immune-mediated adverse reactions occurred at an incidence of <1% (unless otherwise noted) in patients who received KEYTRUDA or were reported with the use of other antiPD-1/PD-L1 treatments. Severe or fatal cases have been reported for some of these adverse reactions. Cardiac/Vascular: Myocarditis, pericarditis, vasculitis; Nervous System: Meningitis, encephalitis, myelitis and demyelination, myasthenic syndrome/myasthenia gravis (including exacerbation), Guillain-Barr syndrome, nerve paresis, autoimmune neuropathy; Ocular: Uveitis, iritis and other ocular inflammatory toxicities can occur. Some cases can be associated with retinal detachment. Various grades of visual impairment, including blindness, can occur. If uveitis occurs in combination with other immune-mediated adverse reactions, consider a Vogt-Koyanagi-Harada-like syndrome, as this may require treatment with systemic steroids to reduce the risk of permanent vision loss; Gastrointestinal: Pancreatitis, to include increases in serum amylase and lipase levels, gastritis, duodenitis; Musculoskeletal and Connective Tissue: Myositis/polymyositis rhabdomyolysis (and associated sequelae, including renal failure), arthritis (1.5%), polymyalgia rheumatica; Endocrine: Hypoparathyroidism; Hematologic/Immune: Hemolytic anemia, aplastic anemia, hemophagocytic lymphohistiocytosis, systemic inflammatory response syndrome, histiocytic necrotizing lymphadenitis (Kikuchi lymphadenitis), sarcoidosis, immune thrombocytopenic purpura, solid organ transplant rejection.

Infusion-Related Reactions

KEYTRUDA can cause severe or life-threatening infusion-related reactions, including hypersensitivity and anaphylaxis, which have been reported in 0.2% of 2799 patients receiving KEYTRUDA. Monitor for signs and symptoms of infusion-related reactions. Interrupt or slow the rate of infusion for Grade 1 or Grade 2 reactions. For Grade 3 or Grade 4 reactions, stop infusion and permanently discontinue KEYTRUDA.

Complications of Allogeneic Hematopoietic Stem Cell Transplantation (HSCT)

Fatal and other serious complications can occur in patients who receive allogeneic HSCT before or after antiPD-1/PD-L1 treatment. Transplant-related complications include hyperacute graft-versus-host disease (GVHD), acute and chronic GVHD, hepatic veno-occlusive disease after reduced intensity conditioning, and steroid-requiring febrile syndrome (without an identified infectious cause). These complications may occur despite intervening therapy between antiPD-1/PD-L1 treatment and allogeneic HSCT. Follow patients closely for evidence of these complications and intervene promptly. Consider the benefit vs risks of using antiPD-1/PD-L1 treatments prior to or after an allogeneic HSCT.

Increased Mortality in Patients With Multiple Myeloma

In trials in patients with multiple myeloma, the addition of KEYTRUDA to a thalidomide analogue plus dexamethasone resulted in increased mortality. Treatment of these patients with an antiPD-1/PD-L1 treatment in this combination is not recommended outside of controlled trials.

Embryofetal Toxicity

Based on its mechanism of action, KEYTRUDA can cause fetal harm when administered to a pregnant woman. Advise women of this potential risk. In females of reproductive potential, verify pregnancy status prior to initiating KEYTRUDA and advise them to use effective contraception during treatment and for 4 months after the last dose.

Read more:
FDA Approves Merck's KEYTRUDA (pembrolizumab) in Combination With Chemotherapy for Patients With Locally Recurrent Unresectable or Metastatic...

Read More...

Transfection Technologies Market Size Research Reports Global Industry, Share, In-Depth Qualitative Insights, Explosive Growth Opportunity|| Lonza,…

Tuesday, November 17th, 2020

DBMR announces the release of the reportTransfection Technologies MarketSize, Share & Trends Analysis Report By 2027. An all-inclusive Transfection Technologies report contains a chapter on the global market and all its associated companies with their profiles, which gives valuable data pertaining to their outlook in terms of finances, product portfolios, investment plans, and marketing and business strategies. The report helps to achieve a dream of an outshining and winning business. This Transfection Technologies market research report helps in answering many business challenges more quickly and saves a lot of time. Moreover, the report consists of all the detailed profiles for the Transfection Technologies markets major manufacturers and importers who are influencing the market.

Global transfection technologies market is registering a substantial CAGR of 9.74% in the forecast period of 2019-2026.

Download Free Sample (350 Pages PDF) Report: To Know the Impact of COVID-19 on this Industry @https://www.databridgemarketresearch.com/request-a-sample/?dbmr=global-transfection-technologies-market&ab

Market Definition

Transfection is an approach to produce exogenous nucleic acids such as DNA, RNA or oligonucleotide into cells. Such nucleic acids can be transferred by polymeric or lipid transfection reagents which promote the cellular absorption. This method is widely used for genomic studies (cell representation, testing, RNA interference, in vitro research) but can be conducted for bio-production (vaccine and protein manufacturing) or medicinal reasons (animal cell treatment). Nucleic acid delivery to cells can be accomplished by distinct physical techniques, such as electroporation, sonoporation or microinjection; however, these procedures are comparatively hazardous to cells. Transfection with chemical substances is a better option for maintaining healthy cell feasibility.

Global Transfection Technologies Market 2020 Report encompasses an infinite knowledge and information on what the markets definition, classifications, applications, and engagements are and also explains the drivers and restraints of the market which is obtained from SWOT analysis. By applying market intelligence for this Transfection Technologies Market report, industry expert measure strategic options, summarize successful action plans and support companies with critical bottom-line decisions. Additionally, the data, facts and figures collected to generate this market report are obtained forms the trustworthy sources such as websites, journals, mergers, newspapers and other authentic sources. Development policies and plans are discussed as well as manufacturing processes and cost structures are also analyzed. This report also states import/export consumption, supply and demand Figures, price, cost, revenue and gross margins.

TO UNDERSTAND HOW COVID-19 IMPACT IS COVERED IN THIS REPORT, GET FREE COVID-19 SAMPLE @https://www.databridgemarketresearch.com/covid-19-impact/global-transfection-technologies-market?ab

List of Companies Profiled in the Transfection Technologies Market Report are:

LonzaPromega CorporationSigma-Aldrich Co.Thermo Fisher Scientific IncBio-Rad Laboratories,Roche Molecular SystemsQIAGENInovio PharmaceuticalsPOLYPLUS TRANSFECTIONComplete Report is Available (Including Full TOC, List of Tables & Figures, Graphs, and Chart) @https://www.databridgemarketresearch.com/toc/?dbmr=global-transfection-technologies-market&ab

Transfection Technologies Report displays data on key players, major collaborations, merger & acquisitions along with trending innovation and business policies. The report highlights current and future market trends and carries out analysis of the effect of buyers, substitutes, new entrants, competitors, and suppliers on the market. The key topics that have been explained in this Transfection Technologies market report include market definition, market segmentation, key developments, competitive analysis and research methodology. To accomplish maximum return on investment (ROI), its very essential to be acquainted with market parameters such as brand awareness, market landscape, possible future issues, industry trends and customer behavior where this Transfection Technologies report comes into play.

The Segments and Sub-Section of Transfection Technologies Market are shown below:

Segmentation: Global Transfection Technologies Market

By Transfection Method

Cotransfection

Electroporation

Cationic Lipid Transfection

In Vivo Transfection

By Applications

Virus Production

Protein Production

Gene Silencing

Stem Cell Reprogramming & Differentiation

Stable Cell Line Generation

Market Size Segmentation by Re gion & Countries (Customizable):

North America (Canada, United States & Mexico)

Europe (Germany, the United Kingdom, BeNeLux, France, Russia & Italy)

Asia-Pacific (Japan, South Korea, China, India & Southeast Asia)

South America (Argentina, Brazil, Peru, Colombia, Etc.)

Middle East & Africa (United Arab Emirates, Egypt, Saudi Arabia, Nigeria & South Africa)

For More Information or Query or Customization Before Buying,Visit@https://www.databridgemarketresearch.com/inquire-before-buying/?dbmr=global-transfection-technologies-market&ab

Market Drivers

Surge in research & development in the field of cell based therapies is contributing to the growth of the market

Massive funds by government and private players is boosting the growth of the market

Growing occurrences of cancer diseases is propelling the growth of the market

Increasing number of obese and overweight population is driving the growth of the market

Market Restraints

Cost of transfection technology instruments is hampering the growth of the market

Hazard of negative reaction with the cell is hindering the growth of the market

Home brew reagents restricts sale of business supply which is restricting the growth of the market

Strategic Points Covered in Table of Content of Global Transfection Technologies Market:

Chapter 1: Introduction, market driving force product Objective of Study and Research Scope the Transfection Technologies market

Chapter 2: Exclusive Summary the basic information of the Transfection Technologies Market.

Chapter 3: Displaying the Market Dynamics- Drivers, Trends and Challenges of the Transfection Technologies

Chapter 4: Presenting the Transfection Technologies Market Factor Analysis Porters Five Forces, Supply/Value Chain, PESTEL analysis, Market Entropy, Patent/Trademark Analysis.

Chapter 5: Displaying market size by Type, End User and Region 2010-2019

Chapter 6: Evaluating the leading manufacturers of the Transfection Technologies market which consists of its Competitive Landscape, Peer Group Analysis, BCG Matrix & Company Profile

Chapter 7: To evaluate the market by segments, by countries and by manufacturers with revenue share and sales by key countries (2020-2027).

Chapter 8 & 9: Displaying the Appendix, Methodology and Data Source

Finally, Transfection Technologies Market is a valuable source of guidance for individuals and companies in decision framework.

Thanks for reading this article; you can also get individual chapter wise section or region wise report version like North America, Europe or Asia.

See the article here:
Transfection Technologies Market Size Research Reports Global Industry, Share, In-Depth Qualitative Insights, Explosive Growth Opportunity|| Lonza,...

Read More...

Exosome Diagnostics And Therapeutics Market Worldwide Industry Analysis and New Market Opportunities Explored – The Daily Philadelphian

Tuesday, November 17th, 2020

Theglobal market for exosome diagnostics and therapeuticsshould grow from $34.7 million in 2018 to $186.2 million by 2023, with a compound annual growth rate (CAGR) of 39.9% for the period of 2018-2023.

Report Scope:

This report represents a current and important business tool to evaluate new commercial opportunities in the exosome diagnostic, therapeutic and research tool markets. The geographic scope of this study covers the U.S. and companies worldwide. This market is complex and consists of a number of different sectors, each affected differently by scientific and technological development. The report identifies the main positive and negative factors in each sector and forecasts further trends and product and assay development in every category of this industry.

Get Access to sample pages @https://www.trendsmarketresearch.com/report/sample/11684

Report Includes:

106 tables An overview of the global market for exosome diagnostics and therapeutics Analyses of global market trends, with data from 2017, 2018, and projections of compound annual growth rates (CAGRs) through 2023 Description of immunological compatibility, cargo capabilities and other intrinsic therapeutic activities of exosomes Details of isolation and detection techniques and description of reagents and tools used for exosome research Evaluation of exosomal proteins and nucleic acids as diagnostic biomarkers and discussion of their impact on microRNA, liquid biopsy and stem cell research industry Information on bioinformatics databases for exosome research and product development, including as ExoCarta, Vesiclopedia and EVpedia Company profiles of the prominent players, including Codiak BioSciences, Evox Therapeutics Ltd., Exosome Diagnostics Inc., HansaBioMed Life Sciences Ltd. (Lonza), NonoSomiX Inc., and System Biosciences (SBI)

Summary

Initially, exosome particles were considered garbage molecules secreted by cells. Today, many researchers are convinced that these tiny vesicles have unlimited potential in diagnostics and therapeutics, especially in oncology treatments.

By definition, exosomes are small membrane sacs/vesicles, approximately 30 to 100 nanometers (nm) in diameter, that are released by both healthy and cancerous cells. Substances from cell cytoplasmsuch as genomic DNA, various RNA species, proteins and lipidsare encapsulated into exosomes and shed into the extracellular environment.

Research shows that all fluids in the human body contain exosomes, which can transfer cytoplasmic ingredients to other cells either locally or at distant sites. Once they reach the recipient cells, cytoplasmic ingredients can alter their biology. Thus, exosomes are widely being adopted by several end users, including hospitals, diagnostics centers and research institutions.

Scientists believe that various biomolecules in exosomes can be profiled and, consequently, may serve as useful biomarkers for different diseases. Nucleic acids such as RNA or DNA can be isolated from exosomes and further analyzed by various techniques. This is now efficiently done with the use of software provided by key players. The software used in exosome therapeutic and diagnostic applications are used for extraction, isolation and other purposes.

Overall, the market for the exosome approach can be divided into three main segments: product, application and end user. The overall market is estimated to be worth $REDACTED million (see Summary Table below), with the potential to increase to $REDACTED million in the next five years with a compound annual growth rate (CAGR) of REDACTED%.

This report evaluates the diagnostic market affected by exosome research and the further potential of exosome-based tests and assays. Indeed, an exosome approach represents the opportunity to expand and develop the liquid biopsy market, a growing sector in cancer diagnostics. Thus, increasing cancer prevalence worldwide generates huge opportunities for the liquid biopsy market. As per Our World in Data, about 42 million people worldwide had cancer in 2016, a more than two-fold increase from 1990. This report also highlights developments in therapeutic and drug development sectors. There is a significant potential for using exosome depletion as a way of treating disease; cancer-generatedexosomes can inhibit the immune response and stimulate angiogenesis, the development of new blood vessels. Consequently, if these exosomes are removed, tumor growth might be inhibited, and anticancer agents can work more efficiently.

In addition, there is a potential for exosomes to be used as targeted delivery vehicles of therapeutic molecules to cancer cellsfor example, delivering small interfering RNA-specific molecules (siRNA) for a particular oncogene expressed in a tumor cell. As per the U.S. National Library of Medicine, cell-based exosomes have a large number of roles and targets. Also, various studies have shown that exosomes have the potential to deliver various types of cargo to target cells efficiently.

Original post:
Exosome Diagnostics And Therapeutics Market Worldwide Industry Analysis and New Market Opportunities Explored - The Daily Philadelphian

Read More...

Page 5«..4567..1020..»


2024 © StemCell Therapy is proudly powered by WordPress
Entries (RSS) Comments (RSS) | Violinesth by Patrick