header logo image


Page 6«..5678..2030..»

Archive for the ‘Death by Stem Cells’ Category

Epigenetic Changes Implicated in Age-related Diminution in Vision and Its Possible Reversal – JD Supra

Wednesday, December 16th, 2020

The Fountain of Youth -- an enduring aspiration, particularly as the ravages of age reduce human faculties prior to leading inexorably to death. Reduction in sight is the human faculty that can have the greatest effect on quality of life in the aged -- a faculty that begins to decline in the 4th or 5th decade of life and doesn't get better (when it does) without medical intervention.

But what if there were a way to rejuvenate sight? That prospect is the tantalizing suggestion in a paper published on December 2nd entitled "Reprogramming to recover youthful epigenetic information and restore vision," Nature 588: 124-29*. The basis of the report is the recognition that many of the age-related effects on vision are an example of gene expression differences associated with epigenetic changes in chromosomal DNA. Epigenetics is a phenomenon of gene structure and expression involving small differences in nucleotide bases, typically methylation of cytosine residues at specific (CpG) sites. These changes have been studied in normal development, where gene expression changes arise as different cell types properly differentiate and act as a molecular "clock" reflecting age. The ability to turn back cellular time has been demonstrated by the development of induced pluripotent stem cells (iPSCs), wherein terminally differentiated somatic cells (typically fibroblasts) can be turned into pluripotent cells. Pluripotent cells are capable of differentiating into cells of each embryonic germinal layer (ectoderm, mesoderm, endoderm), and iPSCs can be produced by expressing four specific genes: OCT4, SOX2, KLF4 and MYC. All of these genes encode transcription factors capable of affecting (and effecting) developmentally relevant gene expression. Consequent to this "de-differentiation" occasioned by expression of these genes is a "resetting" of the epigenetic patterns associated with development. In this paper the researchers hypothesized that resetting these epigenetic patterns could also rejuvenate neuroretinal cells to reinvigorate and overcome the ocular nerve damages by glaucoma in an animal model.

Because one of these genes (MYC) is also associated with cancer development (i.e., it is an oncogene) the researchers developed an inducible expression construct that expressed only the OCT4, SOX2, and KLF4 members of the quartet (OSK). (This decision was also informed by the experience of other researchers that continuous expression of all four genes in animal models resulted in teratomas or was fatal within days of introduction.) Their system used a polycistronic (i.e., all the genes in one linear array) construct of all three genes regulated by a tetracycline response element (TRE) promoter in a adeno-associated viral vector. This construct was tested by introduction into fibroblasts from aged (20 month old) mice and gene expression related to aging (i.e., that showed differential expression with age) was evaluated. These studies showed that OSK expression for 5 days resulted in a "youthful" mRNA expression pattern in these genes (without any effect on the terminal differentiation state of the fibroblasts).

The TRE promoter enabled selection for or against expression of the OSK gene cassette; as the authors explain "[t]he TRE promoter can be activated either by reverse tetracycline-controlled transactivator (rtTA) in the presence of the tetracycline derivative doxycycline (DOX) ('Tet-On') or by tetracycline-controlled transactivator (tTA) in the absence of DOX ('Tet-Off')." Simply put, the presence of absence of DOX in the animal's drinking water determined whether the expression cassette is "on" or "off," as illustrated in this figure:

Long-term (10-18 months) expression of this cassette was achieved in both young (5 months-old) and aged mice with no tumorigenesis or other negative side effects being observed.

To test the ability of induced OSK expression to rejuvenate optical nerve cells the researchers examined retinal ganglion cells (RGC, which project axons away from the retina informing the optic nerve) in an optic nerve crush injury model (which mimics the effects of optic nerve injury due to inter alia glaucoma). The construct was delivered by injection into the vitreous humor and resulted in about 37% of the RGCs taking up and expressing the OSK genes in response to DOX administration. A separate cohort of mice were administered versions of the construct where DOX inhibited OSK expression. In these experiments, "the greatest extent of axon regeneration and RGC survival occurred when all three genes were delivered and expressed as a polycistron within the same AAV particle" according to the researchers. In contrast, inhibition of OSK expression in the "Tet-Off" mice showed no axonal growth. Moreover, delivery of the OSK genes individually in separate viral vectors or in pairs also did not show axonal growth, indicating the need for these genes to be expressed together in proper relative amounts provided by the polycistronic construct. The researchers also found OSK expression induced expression of Stat3, a gene know to encourage regeneration. These results were obtained in using 12-month-old mice as well as 1- and 3-month-old mice, which indicated, as the authors note, that "ageing does not greatly diminish the ability of OSK transcription factors to induce axon regeneration." Increased axonal growth from RGCs was found even after crush injury, an effect found with no other treatment modalities.

The researchers then determined whether these reinvigorated RGCs showed changes in DNA methylation patterns. In the absence of DOX-induced OSK expression injury in this model caused an "accelerated" aging pattern, whereas in the presence of DOX-induced OSK expression counteracted this effect according to the results reported in this paper. Interestingly, this preservation of a "youthful" pattern of DNA methylation was found to be enriched at genes "associated with light detection and synaptic transmission." Having shown this association the researchers then investigated whether axonal regeneration required youthful changes in DNA methylation. These experiments were performed by reducing expression of genes that caused DNA demethylation in RGCs (and whose expression was known to be increased in cells expressing OSK) and detecting that axonal regeneration did not occur in these mice even in the presence of DOX-induced OSK expression.

Whether these effects of OSK expression would also be seen in human neurons was investigated using differentiated human neurons in vitro. Neurons harboring an OSK-encoding construct were treated with vincristine (a drug that occasions axon injury) and DOX-induced OSK expression was shown to "counteract[] axonal loss and the advancement of DNA methylation age," showing a 15-fold greater area of proliferation in OSK-expressing cells than control vincristine-treated neural cells. These cells also showed the demethylation-dependent characteristics that were shown in RGCs in the mouse optic nerve crush injury model.

The most clinically significant result disclosed in this paper involved the effect of OSK expression in a glaucoma model in vivo. Intraocular pressure was increased to pathological levels by injecting microbeads unilaterally into the anterior chamber of mouse eye for 21 days. At 4 weeks, after these animals showed correspondingly unilateral decreases in axonal density and the number of RGCs present in the treated eye. The viral vector encoding inducible OSK expression thereafter was introduced by intravitreal injection followed by DOX-induced OSK expression for 4 weeks. Compared with control (introduction of saline or viral vectors not encoding OSK into the microbead-treated eyes) the OSK vector-treated eyes showed "restored axon density equivalent to that in the non-glaucomatous eyes, with no evidence of RGC proliferation." These mice also showed a reversal of vision loss caused by the glaucomatous injury. Together these results indicated that OSK expression could be a therapy for glaucoma in humans.

Finally, the paper reports efforts to determine whether OSK expression could improve age-related (as opposed to injury- or pathology-related) vision problems. In these experiments, 3-and 11-month-old mice were treated by intravitreal injection of DOX-inducible OSK encoding constructs and OSK expression induced for 4 weeks. Twelve-month-old mice showed age-related visual acuity and RGS electrical activity diminution which was reversed by DOX-induced OSK expression. However, these phenotypic changes were not observed to be associated with an increased number of RGCs or axon density, which prompted these researchers to hypothesize that the effect were dependent on changes in gene expression ("transcriptomic changes" as these were termed in the paper). RGCs from treated or untreated 12-month-old mice were isolated and compared with RGCs from 5-month-old mice and expression of 464 genes were found to be altered: expression of almost all (90%) of these genes were found to be restored to youthful levels in OSK-expressing RGCs. The participation of DNA methylation changes in aged RGCs in producing a youthful pattern of gene expression was further assessed and validated using artificial intelligence/machine learning approaches.

The results reported in this paper suggest therapeutic interventions that could improve vision in the aged human population even in the absence of vision-impairing pathologies such as glaucoma. Although cautious to mention that "we do not wish to imply that DNA methylation is the only epigenetic mark involved in this process" and "[i]t is likely to involve other transcription factors and epigenetic modifications," the authors are not blind to the implication that:

[W]e show that it is possible to safely reverse the age of a complex tissue and restore its biological function in vivo. Using the eye as a model system, we present evidence that the ectopic expression of OSK transcription factors safely induces in vivo epigenetic restoration of aged CNS neurons, without causing a loss of cell identity or pluripotency. Instead, OSK promotes a youthful epigenetic signature and gene-expression pattern that causes the neurons to function as though they were young again. The requirement for active demethylation in this process supports the idea that changes in DNA methylation patterns are involved in the ageing process and its functional reversal.

* By researchers from Harvard Medical School, Yale University School of Medicine, Massachusetts General Hospital, UCLA Geffen School of Medicine, and The University of New South Wales Medical School.

View post:
Epigenetic Changes Implicated in Age-related Diminution in Vision and Its Possible Reversal - JD Supra

Read More...

NASA-partnered Pluristem crashes to Earth as it axes leading therapy – FierceBiotech

Wednesday, December 16th, 2020

Israeli biotech Pluristem is canning its experimental phase 3 critical limb ischemia therapy after an outside review said it was no good.

Haifa, Israel-based Pluristems R&D operation is built upon placenta-derived adherent stromal cells, which the biotech has designed for use in patients of all human leukocyte antigen types. This approach is made possible by the low immunogenicity of the cells. Once inside the body, Pluristem hopes the cells will drive the healing of injured tissue.

But one of its leading contenders using this approach has been judged a failure in phase 3: An independent data monitoring committee (DMC) took a look at the ongoing data for its pivotal phase 3 in patients with critical limb ischemia (CLI), a severe obstruction of the arteries which markedly reduces blood flow to the extremities and can lead to amputation.

Your Guide to Maximizing the Benefits of a Unified Clinical Data Capture and Management Platform

Learn how a unified platform can increase visibility across your trial. Adopt technology solutions driving faster, more accurate data interpretation for better decision-making to remain competitive in a new era of drug development.

The DMC said the test was unlikely to meet the primary endpoint, and that the CLI study population has experienced a substantial low number of events (major amputation of the index leg or death), different from what is known in clinical medicine for the rate of these events in this patient population. The lower than anticipated event rate in the placebo group reduced the statistical power of the study to meet its primary endpoint.

The biotech is now tossing out the therapy and will instead focus on other pipeline areas, including a long-shot stem cell attempt at treating COVID-19. The biotechs shares fell nearly 40% on the news.

We are deeply disappointed by the outcome of the CLI interim analysis. In light of the DMCs recommendation, we decided that it would be in the best interests of the company and its shareholders to terminate the CLI study and focus our resources and efforts on our other lead indications, said Pluristem CEO and President Yaky Yanay.

We expect to present topline clinical results during calendar year 2021, including our phase 3 study in muscle regeneration following hip fracture, phase 2 studies in Acute Respiratory Distress Syndrome associated with COVID-19 and our phase 1 study in incomplete hematopoietic recovery following hematopoietic cell transplantation. Pluristem is well positioned to advance and support future development of these indications.

Last year, Pluristem penned a deal with NASA to assess its cell therapies against the health problems caused by spending time in space, teaming up with NASAs Ames Research Center for the project, which focuses on using its PLX placenta-derived cell therapies to try to prevent or treat medical conditions that can occur during and after space missions, including conditions that affect the blood, bone, muscle, brain and heart.

See the original post here:
NASA-partnered Pluristem crashes to Earth as it axes leading therapy - FierceBiotech

Read More...

Exploring Increase of Point Mutations Associated with High-Dose Melphalan in Multiple Myeloma – Targeted Oncology

Wednesday, December 16th, 2020

High-dose melphalan (Evomela) followed by autologous stem cell transplant (ASCT) has demonstrated significant efficacy for the treatment of patients with multiple myeloma. The agent has improved progression-free survival (PFS) in patients when administered as upfront therapy to patients aged 66 years or younger.

The median PFS shown with melphalan plus ASCT in a phase IFM/DFCI 2009 study was 50 months versus 26 months when compared with the standard of care treatment regimen lenalidomide (Revlimib), bortezomib (Velcade), and dexamethasone (RVD) alone hazard ratio for disease progression or death, 0.65;P<.001). As the treatment strategy continues to be explored in patients with multiple myeloma, researchers have now begun to investigate an outstanding biological question of whether the alkylating agent causes an increased amount of DNA damage.

An analysis presented during the virtual 2020 American Society of Hematology (ASH) Annual Meeting showed that between the time of diagnosis and relapse, patients treated with high-dose melphalan had an increased number of point mutations. It was unclear from this research how this result translated to treatment selection and sequencing.

In an interview with Targeted Oncology, Mehmet Samur, PhD, senior research scientist, Dana-Farber Cancer Institute, discussed the investigation of high-dose melphalan following ASCT in patients with multiple myeloma and shared insights into how the ongoing questions can be explored in the future.

TARGETED ONCOLOGY: Can you explain what was demonstrated prior with high-dose melphalan followed by ASCT in patients with multiple myeloma?

Sumar: The clinical part of the phase 3 study was published previously. It showed that when you do RVD plus high-dose melphalan following by stem cell treatment, patients do significantly better than patients who get RVD alone. Adding high-dose melphalan increased the PFS benefit by around 12 months.

TARGETED ONCOLOGY: Can you provide background on your analysis of Melphalan for patients with multiple myeloma?

Sumar: Melphalan is an alkylating agent. Because of the way the agent works, we always think that it creates more DNA damage. The study that we presented at ASH was questioning whether this was true or not.

We collected DNA sequencing data from patients who were treated with RVD followed by high-dose Melphalan and a bone marrow transplant. We had a total of 25 patients, and we collected data at the time of diagnosis and the time of relapse. To compare this compilation, we also collected data from 43 patients from the IFM/DFCI 2009 study who only received RVD. We also collected data at diagnosis and relapse in the 43 patients. Genomic alterations were compared at diagnosis and relapse for patients who were injected with high-dose Melphalan and RVD versus patients who were only treated with RVD.

TARGETED ONCOLOGY: What were the findings from this study?

Sumar: We found that patients who got high-dose melphalan plus RVD followed by transplant accumulated more point mutations. To be precise, they accumulated around 10,000 new mutations between diagnosis and relapse at 5 years. For RVD patients, there were around 4500 new point mutations. The study showed that treating patients with high-dose Melphalan is increasing the mutational load by about 2.9-fold at the time of relapse.

TARGETED ONCOLOGY: What are the implications of these findings?

Sumar: There are a couple of things that we see from our study. One point is that we only saw point mutations. We didnt see any large-scale DNA alterations. This suggests that our patients who are treated with high-dose melphalan are more likely to experience changes.

In terms of the pathways that are mutated, the DNA damage repair pathway is more frequently mutated between diagnosis and relapse in patients treated with high-dose melphalan. We think that if we combine inhibitors that can overcome the selection of DNA damage repair pathway mutations, those patients may get additional benefit from the treatment.

We dont have clear data yet on whether this increased mutational load is something that is bad for patients. Even though these patients have more mutations, overall survival times are similar between the 2 arms. C outcomes are not impacted by the increased number of mutation so far.

TARGETED ONCOLOGY: What plan are underway to further this research?

Sumar: We are expanding our study in multiple ways. There is no clear data set we can get answers to our ongoing questions yet. We have reached out to our partners around the world to see if we can come up with a cohort to investigation. Also, we are looking at impact of these mutational load increase on other features like secondary cancer rate.

TARGETED ONCOLOGY: The understanding of gene mutations in myeloma is evolving. Can you discuss the current role of genomic testing?

Sumar: It has been shown in many studies that genetic testing at diagnosis can tell us which patients are high risk and which are low risk. Studies have also shown that patients who have loss of p53 or with deletion 17p will have bad outcomes.

There was a study published last year in the Journal of Clinical Oncology showing which patients with myeloma would have a lower risk. The study also shows that there are certain genomic features prolong survival time in patients.

We have different genomic tools that we can use to look at these different alterations and assess patient risk. Today, I think people are looking at these alterations from all different angles to plan stratification in upcoming clinical trials.

TARGETED ONCOLOGY: In your opinion, what change will we see in the myeloma treatment landscape in the next 5 years?

Sumar: There are a lot of studies looking at new treatment. Everyone is carefully watching out for data on new treatment options like chimeric antigen receptor T-cell therapy, bispecific antibodies, and monoclonal antibodies. It looks like these agents are providing benefit to patients, but they are at the very early stages of research.

Reference:Attal M, Lauwers-Cances V, Hulin C, et al. Lenalidomide, bortezomib, and dexamethasone with transplantation for myeloma. N Engl J Med. 2017; 376(14):1311-1320. doi: 10.1056/NEJMoa1611750

View post:
Exploring Increase of Point Mutations Associated with High-Dose Melphalan in Multiple Myeloma - Targeted Oncology

Read More...

2020 at the U: The year in review – University of Miami

Wednesday, December 16th, 2020

Take a look at a month-by-month recap of the biggest stories at the University of Miami during the past year.

From a pandemic that forced the migration of spring semester classes to an online environment, to a new dean for the College of Engineering, to a Super Bowl halftime performance by the Band of the Hour, to a record-breaking gift for the Sylvester Comprehensive Cancer Center, 2020 proved to be a very unusual year for the University of Miami.

January

The Planet Kreyol student organization and the Office of Multicultural Student Affairs commemorate victims of the 2010 Haiti earthquake with dance, song, poetry, and more on January 15.

With evidence mounting that climate change is worsening everything from wildfires to hurricanes, the University of Miamis Rosenstiel School of Marine and Atmospheric Science hosts the three-day Miami Climate Symposium 2020: Predicting and Living with Extremes. The summitheld Jan. 22-24explored how sea level rise, saltwater intrusion, extreme heat waves, and other conditions exacerbate tropical cyclones, storm surge, and coastal flooding, as well as analyzed adaptation policies and strategies.

A professor of jazz trumpet at the Frost School of Music and four alumni of the University of Miami receive Grammy Awards during the 62nd annual ceremony held Jan. 26 in Los Angeles. Brian Lynch, jazz trumpet professor, is honored in the category of Best Large Jazz Ensemble. Cristian Macelaru, B.M. 03; Julio Reyes Copello, M.M. 00; Carlos Fernando Lopez, B.M. 12; and Natalia Ramirez, M.A. 17, also win Grammy Awards.

Miami baseball alumnus Tommy Adams makes the largest gift in support of the baseball program by a former University baseball student-athlete, donating $1 million toward the University of Miamis Baseball Facility Enhancement Campaign.

February

Five months after Hurricane Dorian devastated the Bahamas, students enrolled in the semester-long action project interdisciplinary class offered by the Miami Herbert Business School travel to Freeport for a three-day visit, meeting with Bahamian students and local businesses to share ideas and inspire hope.

Building on the immense resources and expanding the vision of the Center for Computational Science, the University establishes the Miami Institute for Data Science and Computing to catalyze data-intensive research that will solve real-world problems and enhance the understanding of data science among students and the public.

The Lancet, one of the worlds leading medical journals, announces that Felicia Marie Knaul, director of the University of Miami Institute for Advanced Study of the Americas, will lead a new Lancet Commission examining gender-based violence and maltreatment of young people, two areas with a dearth of study and understanding.

Students from the University of Miamis Frost Band of the Hour and the color guard, as well as the Hurricanettes dancers, perform in the Pepsi Super Bowl LIV Halftime Show at Hard Rock Stadium on Feb. 2.

Redshirt senior David Dinsmore wins his fourth straight gold medal in the mens platform on Feb. 21, capturing top honors at the 2020 ACC Swimming and Diving Championships.

At the Dolphins Cancer Challenge, Team Hurricaneswith 1,250 strongjoined thousands of others on Feb. 29 to run, walk, and ride to fight cancer and support the Sylvester Comprehensive Cancer Center.

March

For the health and well-being of the campus community, the University extends spring break for students through March 22, announcing that classes will resume on March 23 but strictly in online environments through at least April 4. Shortly thereafter, with COVID-19 cases surging across the nation, the University, in accordance with public health guidance to reduce density on campus, extends online instruction through the remainder of the spring semester and implements partial closing of on-campus housing.

April

An international team of scientists led by Dr. Camillo Ricordi, director of the Diabetes Research Institute and Cell Transplant Center at the University of Miami Miller School of Medicine, is granted immediate FDA authorization for a 24-patient clinical trial to test the safety and exploratory efficacy of umbilical cord-derived mesenchymal stem cells to block the life-threatening lung inflammation that accompanies severe cases of COVID-19.

Joined by the Rapid Defense Network in New York, the Southern Poverty Law Center, and others, the School of Laws Immigration Clinic files a lawsuit on April 13 accusing U.S. Immigration and Customs Enforcement authorities of ignoring COVID-19 guidelines in three Florida detention centers.

Debbie Ajagbe is named the 2020 ACC Indoor Track Scholar-Athlete of the Year on April 17, with five other Miami women joining her on the All-ACC Academic Team for Indoor Track and Field. Earlier in the year, Ajagbe, a mechanical engineering major, earned both ACC Womens Field Performer of the Year and ACC Championship Field MVP honors, winning both the weight throw and shot put at the conference championships.

May

The 180 graduates of the Miller School of Medicines Class of 2020 celebrate their newly minted degrees during a virtual commencement on May 9.

Arva Moore Parks, a prominent historian and preservationist, who served on the University of Miami Board of Trustees for 26 years and wrote several books on Greater Miami, Coral Gables, and University history, passes away on May 10.

Nine University of Miami Athletics programsmens basketball, mens and womens cross country, mens diving, golf, rowing, mens and womens tennis, and womens track and fieldare recognized for perfect single-year scores of 1,000 in the 2018-19 Academic Progress Report released May 19 by the NCAA.

UMTVs first Black show, The Culture, is nominated by the Suncoast Chapter of the National Academy of Television Arts & Sciences for a student production award in the magazine program category.

Brian Van Belle and Chris McMahon, two of the Miami Hurricanes best pitchers, are named All-Americans by Collegiate Baseball Newspaper on May 26. Both Van Belle and McMahon earned spots on the second team after posting brilliant performances in the abbreviated 2020 season that was halted due to the COVID-19 pandemic.

June

Pratim Biswas, the Lucy and Stanley Lopata Professor in the McKelvey School of Engineering at Washington University in St. Louis and a pioneer in his field recognized for applying aerosol science and engineering to multiple areas, is named dean of the University of Miami College of Engineering.

The University of Miami Board of Trustees elects six new members to its ranks with expertise in business, finance, law, technology, and strategic planning. Patricia Menendez-Cambo, Adam E. Carlin, Jose R. Mas, Alice S. Vilma, Carolyn B. Lamm, and Jordan Rhodes were elected to the Board on June 19.

Dr. Judy Schaechter, chair of the Miller School of Medicine Department of Pediatrics, is named a 2020-21 Health Policy Fellow by the Robert Wood Johnson Foundation and the National Academy of Medicine. The prestigious one-year fellowship in Washington, D.C., will enable her to expand her longtime involvement in health policy and child policy at the local and state levels to the federal level.

July

The University becomes one of 89 locations around the nation, and one of only six in Florida, to enroll volunteers for the first Phase 3 clinical trial of a COVID-19 vaccine. The trial, part of the National Institutes of Health COVID-19 Prevention Trials Network, tested a vaccine developed by scientists at the NIHs National Institute of Allergy and Infectious Diseases and collaborators at biotechnology company Moderna, Inc. Vice President Mike Pence visited the Miller School of Medicine along with Florida Gov. Ron DeSantis on July 27 to thank the University for its participation in the trial.

The National Oceanic and Atmospheric Administration selects the Rosenstiel School of Marine and Atmospheric Science to host the Cooperative Institute for Marine and Atmospheric Studies, which will bring together the research and educational resources of 11 partner universities to increase scientific understanding of the Earths oceans and atmosphere within the context of NOAAs mission. The selectionmade through an open, competitive evaluationcomes with an award of up to $310 million over the course of five years, with the potential for renewal for another five years based on successful performance.

Amid ongoing nationwide protests against police brutality sparked by the tragic death of Minneapolis resident George Floyd in May, President Julio Frenk reaffirms his commitment to racial and ethnic equality, outlining in a letter sent to all students, faculty, and staff a 15-point plan the University will implement to support racial equality, inclusion, and justice across the institution and in the greater South Florida community.

Patti Herberta longtime University of Miami alumna and benefactor who, along with her husband Allan, donated millions of dollars to the institution, helping to transform academics and student lifepassed away on Monday, July 27. She was 84.

University of Miami Libraries launches Documenting COVID-19: South Floridas Pandemic Experience. Through community-generated and community-contributed content that will be made available through digital collections and by visiting the library, the initiative will chronicle how local communities are dealing with the crisis.

August

In a move to support a safe learning and working environment for students, faculty, and employees, the Butler Center for Service and Leadership establishes a new team of public health ambassadors to support the Universitys COVID-19 reopening and operating plan. The 75 students who made up the new Public Health Ambassadors Program during the fall semester enforced guidelines on the Coral Gables Campus by offering support and utilizing peer-to-peer influence to encourage members of the campus community to engage in the healthy behaviors of physical distancing, wearing face coverings, and hand washing/sanitizing.

Lakeside Village, a 12-acre facility on the shores of Lake Osceola in the heart of the Coral Gables Campus, officially opens its doors on Aug. 13 to greet the inaugural class of students to the transformative housing complex.

Featuring a mix of virtual and in-person instruction, the first day of fall semester classes begins on Aug. 17. On the Coral Gables Campus, a number of safety protocolsfrom mandatory mask-wearing to social distancinghelp ensure the well-being of students, faculty, and staff.

The Miller School of Medicine Class of 2024 launches the innovative NextGenMD Curriculum, which focuses on health system science and features an enhanced emphasis on mentorships. The students will be significantly better prepared to respond to COVID-19 and to the public health challenges that will follow.

The Miller School of Medicine becomes one of a few medical schools across the nation selected by the National Institutes of Health to test the effectiveness of treating COVID-19 patients with convalescent plasma.

Following a limited opening in April 2020, Canes Central, a new student-centered, service-oriented department, fully opens. It offers both in-person and online undergraduate and graduate students assistance on matters relating to registration and records, billing and payment, financial aid, and Cane Cards.

Legendary Miami Hurricanes baseball coach Jim Morriswho in his 25 seasons at the University of Miami won 1,090 games, made the NCAA postseason 23 straight years, reached the College World Series 13 times, and won national championships in 1999 and 2001is voted into the 2020 induction class of the National College Baseball Hall of Fame.

September

Sylvester Comprehensive Cancer Center at the University of Miami Leonard M. Miller School of Medicine receives a landmark gift of $126 million. The groundbreaking donationthe single largest in the University of Miamis 95-year historywill accelerate breakthrough advances in finding cures for cancer and expand innovative treatment options for cancer patients.

The University of Miami jumps eight spots to No. 49 in U.S. News & World Reports 2021 Best Colleges issue, placing the institution back among the 50 top-tier colleges and universities. This jump in the rankings reflects our commitmenteven in these unprecedented timesto comprehensive excellence and selective preeminence, said President Julio Frenk.

Sylvester Comprehensive Cancer Center, part of the University of Miami Health System, opens the Dwoskin Proton Therapy Center on Sept. 15. The new state-of-the-art facility treats patients with proton therapy, an advanced type of low-dose radiation that is extremely precise and two-thirds the speed of light.

Physician-researchers with the Miller School of Medicine begin a new Phase 3 clinical trial to test another investigational vaccine for COVID-19. Part of a large-scale international trial in partnership with Janssen Pharmaceuticals, the clinical trial to test the Janssen vaccine is the Miller Schools second human study of its kind.

October

The University of Miami becomes the first college testing site for a quick, easy, and cost-effective Israeli-produced COVID-19 breath analyzer that could revolutionize coronavirus testing if approved by the FDA.

In a Miami Herbert Business School webinar held Oct. 8, U.S. Secretary of Health and Human Services Alex Azar highlights the administrations effort to restructure the health care system to combat the COVID-19 pandemic and support countries in Latin America and the Caribbean.

November

Taking their dedication to fighting cancer to a new level, the Miami Dolphins pledge a transformational $75 million gift to Sylvester Comprehensive Cancer Center at the University of Miami Leonard M. Miller School of Medicine, South Floridas only NCI-designated cancer center.

Musicians from the Frost School of Music join Dean Shelly Berg and celebrity musicians to participate in a benefit concert on Thanksgiving Day in support of nurses. The livestreamed Nurse Heroes Live! concert raises funds for the Nurse Heroes Foundation, an initiative working to support and honor nurses.

December

Four extraordinary University of Miami alumniJose R. Mas, Jackie Nespral, Hilarie Bass, and Jaret L. Davisshare their advice with more than 5,000 students at four virtual commencement ceremonies held Dec. 10 and 11.

The rest is here:
2020 at the U: The year in review - University of Miami

Read More...

Something Wrong With Your Sniffer? It Could Be the Coronavirus – Tufts Now

Wednesday, December 16th, 2020

COVID-19 has many symptoms, including fever, coughing, and fatigue. But one of the more distinctive signs is the loss of the ability to smell. Were not talking about the usual stuffy nose that goes along with a cold, but an inability to process scent even when youre not congested. People have reported that not being able to smell their own perfume or finding no aroma in their cup of mint tea was their first clue that they might be infected.

James Schwob, a professor of developmental, molecular, and chemical biology at Tufts University School of Medicine, researches the olfactory system and the roughly 1,000 types of neurons that are involved in our ability to register odors both good and bad.

Right now, he is studying tissue from COVID-19 patients to better understand how the virus leads to anosmia, or loss of sense of smell. Tufts Now talked to Schwob about what we know about viruses and their effects on sense of smell.

Tufts Now: How does sense of smell work?

James Schwob: The sense of smell operates by chemicals wafting in on the air and reaching the upper and back parts of the nasal cavity. Those chemicals bind to receptors on sensory neurons in the epitheliumthe thin tissue that lines the nasal passages. Those neurons then send a signal up the olfactory nerve into the brain, where it registers as the delicious smell of coffee or fresh cut grass.

What causes you to lose it?

There are a number of pretty well-known causes for loss of sense of smell. One is via post-viral infection, and we think that has something to do with the immune system causing inflammation. It also happens with head injuryin that case, its likely the part of the brain that receives the smell signals that is damaged. Toxin exposurefrom cadmium, formaldehyde, or methyl bromide, for examplewill make you lose your sense of smell. Chronic sinus infections and simply the aging process can also cause anosmia.

How common is loss of sense of smell among COVID-19 patients?

We have long known that people can lose their sense of smell after other viral infections, such as the flu, but the percentage of people who have had this problem with COVID-19 is quite remarkable. One study from Iran reported 98 percent of hospitalized patients had an objective problem with their sense of smell.

How does COVID-19 cause people to lose their sense of smell?

We dont really know why this happens with COVID-19. It could be that the virus is harming the neurons that send smell signals to the brain, or that the bodys immune system, in trying to deactivate the virus, is killing other, supporting cells that are part of that pathway.

There is some evidence that certain cells in the lining of the nasal passages express a protein receptor, called ACE2, that the coronavirus uses it as a way to infiltrate the body. That is one of the things Im hoping to investigate.

Back in 2000, colleagues and I published two papers looking at a different coronavirus, called mouse hepatitis virus. We looked at what effect that had on the peripheral olfactory nerve in the central olfactory system. What we found was that this virus would pass up that nerve into the brain and cause problems in the brain.

One of the other things that has been described is that there have been some neurological symptoms due to the infection with the SARS-CoV2-virus. And one of the questions we have is whether the virus is crawling up the nerve in some fashion in these patients who have died of the infection.

The good news is that the olfactory epithelium contains stem cells that can give birth to new neurons throughout life as long as they remain intact. So the system has a capacity to repair itself. Some COVID-19 patients have recovered their sense of smell within a couple weeks. Thats actually quicker than you would expect new neurons to be created, so there could be some sort of functional disruptionrather than neuron deathgoing on.

With all the more serious symptoms associated with COVID-19, why is sense of smell worth investigating?

Sometimes loss of smell is a COVID-19 patients only symptom. Any symptom that can be tied directly to the disease becomes an important one to be aware of, so that it can be used to guide testing and keep people from unknowingly spreading the disease. That is part of the reason I think its important to figure this out.

An intact sense of smell is also critical to good nutrition. If smell is lost so is most of foods flavor. As a consequence, patients may overeat (to try to get the pleasure back), undereat (why bother?) or over-salt or -spice their food, because those aspects of food flavor can still be detected by nerves and taste buds in the oral cavity.

Loss of sense of smell can be very disturbing, because eating is of our great pleasures in life. We dont want to lose that when we have so few pleasures left to us now that were stuck at home.

How can a person know if their sense of smell is really hampered, and they arent just imagining it?

One of the things that can be done pretty easily, pretty objectively by someone at home would be to take some ground coffee and see how far away you can hold it and still smell it. Or do the same with rubbing alcohol or your shampoo. If your nose is not congested and you have trouble recognizing those or other scents that are familiar to you, you might want to call your doctor about getting tested.

Julie Flaherty can be reached at julie.flaherty@tufts.edu.

Link:
Something Wrong With Your Sniffer? It Could Be the Coronavirus - Tufts Now

Read More...

Science can stop us ageing, according to a new book. But do we want it to? – The Independent

Wednesday, December 16th, 2020

I

m going to turn 40 this month, a milestone Im approaching with nine-tenths equanimity, one-tenth mild existential dread.

On the one hand, with a kid and a job, Im too busy to spend much time peering at my newly middle-aged navel. Plus, if youre lucky enough to be healthy, griping about simply growing older is churlish. On the other, there is an undeniable psychic wrench in the realisation that I now tick the aged 40-60 box. Somehow, I seem to have lurched from young and clueless to the age where people will make jokes about the number of candles on my cake, without an intervening period of solid competence. I still run for the bus. I can still never find the lids for the Tupperware. I am older than the prime minister of Finland, but I cant reliably pair my own socks. I dont feel fully grown up, and yet there is the creeping sense that, as Sue Townsend put it via her timeless mouthpiece Adrian Mole, Im on a pathetic slide towards gum disease, wheelchair ramps and death.

But is this slide inevitable? Thats the central question of Ageless: The New Science of Getting Older Without Getting Old, an ambitious and energetic new book by the scientist and writer Andrew Steele. While hes not peddling some holy grail of immortality, he does give a startling round-up of the biological factors that make us age and the emerging techniques to tackle them, offering the prospect of both longer and healthier lives.

To start with some perspective: human lifespans vary hugely based on time and place, with life expectancy under the age of 40 the norm everywhere at the start of the 19th century, largely due to high child mortality. Drawing examples from the animal kingdom, Steele points out that innings vary from five minutes for a mayfly to 400 years for a Greenland shark. We are, of course, neither flies nor sharks. But if we accept the idea that a human life isnt capped at 82 and pursue all the opportunities of frontier research, perhaps we could become more like tortoises. As Steele explains, the Galapagos tortoise, along with a handful of other blessed oddballs in the natural world, are negligibly senescent meaning they have no obvious impairments of movement or senses as they get older, and they experience no age-related declines in fertility.

They dont live forever. But referring to a giant tortoise discovered by Charles Darwin who lived until 2006, Steele writes: Harriet was likely pretty much as sprightly at 170 as she was at 30, at the height of Queen Victorias reign which is to say, not very; she was a giant tortoise, after all.

This liveliness of tone helps to carry the reader through an unsparing account of the toll ageing takes on the human body. The comic writer Nora Ephron may have called her mature memoir I Feel Bad About My Neck, but ageing emerges from this book as less aesthetic challenge, more devastating, systemic collapse. The gloomiest statistic of the lot? Your chance of death doubles every eight years, as a range of processes make us progressively more susceptible to big killers like cancer, heart disease, stroke, dementia and diabetes.

Steele gives a jauntily accessible account of the mechanisms behind our decline. One overarching driver is disposable soma theory: the evolutionary logic that prioritises the health of our reproductive cells over the soma cells which make up our own bodies. I had always suspected that having a child somehow drained me of my life force: here is the evidence. And as our neglected soma cells age, things go wrong. Our telomeres, protective caps on our chromosomes which are essential for healthy cell division, get shorter. Autophagy the process by which cells clear out their own junk declines. Proteins misfold into sticky clumps that risk forming the plaques behind Alzheimers disease. Frail old cells linger: They stick around, no longer dividing aged, zombie cells which refuse to commit cell suicide, known as senescent cells. Our mitochondria which give cells energy misfunction. Stem cells falter. Chronic inflammation ups the risk of everything from diabetes to cancer. Its a startling wake-up call that there are worse things bubbling away in your biology than a few grey hairs, or the frown lines that make every Zoom call of this pandemic a harrowing ordeal for the over-35s.

Independent Culture NewsletterThe best in film, music TV & radio straight to your inbox every week

Independent Culture NewsletterThe best in film, music TV & radio straight to your inbox every week

But what if there was another way? Dont book your return trip to the moon for 2125 just yet. The science has a long way to go: as Steele points out, most of the research is in its early stages, bristling with the potential for pitfalls and unintended consequences. But there are a number of approaches that hold promise, with todays huge advances in computing spurring new therapies. Steele posits that senolytics, or drugs that destroy senescent cells, could be with us in the next few years, albeit to tackle age-related conditions rather than ageing itself. After that, more advanced treatments like gene and stem cell therapies could be available on timescales measured in decades. Ultimately, instead of tackling the individual symptoms of ageing a creaky knee here, a furred-up artery there we will move towards systems medicine that stops us falling to bits in the first place:

The first ageless generation probably wont realise their luck at first theyll grow up expecting to die at 100, or 150, or whatever old is for their society but, one after another, lifesaving medical breakthroughs will push their funerals further and further into the future.

Steele writes with the maverick confidence of the outside perspective. He took a PhD in physics, then moved into computational biology before pursuing his interest in ageing as a writer. In this career switch, he has something in common with Aubrey de Grey, the bearded high priest of the anti-ageing movement who began with a background in computer science. While Steele stops short of making outlandish predictions de Grey famously ruffled academic feathers with his claim people could live to be 1,000 the logic is the same.

While we would all merrily jettison our wrinkles and give bowel cancer a miss, are we really ready for an ageless society, presuming the science stacks up? Steele writes quite reasonably that no one would invent the suffering of old age as a solution to an over-crowded world, but this research nonetheless opens up a fascinating Pandoras box of challenges. Rather than a utopia of dewy-faced centenarians hover-boarding to work, we would probably first see a deepening of the grotesque inequalities that already exist in health today. Theres currently a nine-year gap in lifespans between the richest and poorest parts of the UK. Add some fancy preventive drugs to the mix and you can guess who would get them first. Moreover, with ageism rife in the workplace, especially in the tech industry which provides an increasing number of jobs, who could afford not to quaff the pills that keep you looking fresh in your hoodie?

Writing with the vim of a Bill Bryson and the technical knowledge of a scientist, Steele at least gives us a chance to grasp whats at stake in this dazzling, daunting age where big data meets human biology. Negligible senescence may remain a stretch for my New Years resolutions, but Id happily progress at a more tortoise-like pace towards the next milestones of decrepitude.

Read the rest here:
Science can stop us ageing, according to a new book. But do we want it to? - The Independent

Read More...

Cancer cells destroyed in just 3 days with new technique

Monday, December 7th, 2020

Cancer cells are relentless, possessing the vexatious ability to develop resistance to current therapies and making the disease hugely challenging to treat. However, an exciting new study may have identified cancers weak spot; the discovery has already led to the near-eradication of the disease in cell cultures.

The study which was recently published in the journal Nature Biomedical Engineering reveals how altering the structure of chromatin in cancer cells could make them easier to destroy.

In the cell nucleus, DNA is wrapped around proteins called histones. Together they form chromatin.

Chromatins job is to package the genetic code neatly into the cells nucleus. Chromatin can also regulate which genes are switched on and off. In cancer cells, however, chromatin helps them to evolve and adapt to cancer therapies, thereby allowing them to survive.

If you think of genetics as hardware, explains study co-author Vadim Backman, of the McCormick School of Engineering at Northwestern University in Evanston, IL, then chromatin is the software.

Complex diseases such as cancer, he adds, do not depend on the behavior of individual genes, but on the complex interplay among tens of thousands of genes.

So, Backman and his colleagues set their sights on chromatin as the key to combating cancer drug resistance, and an imaging technique they developed last year helped them to learn more about this intricate set of macromolecules.

The new technique is called Partial Wave Spectroscopic (PWS) microscopy, and it enables real-time monitoring of chromatin in living cells.

Additionally, the researchers explain that PWS allows them to assess chromatin at a length scale of 20200 nanometers, which they say is the precise point at which cancer formation influences chromatin.

They used PWS to monitor chromatin in cultured cancer cells. They found that chromatin has a specific packing density associated with gene expression that helps cancer cells to evade treatments.

The analysis revealed that a more heterogeneous and disordered chromatin packing density was related to greater cancer cell survival in response to chemotherapy. A more conservative and ordered packing density, however, was linked to greater cancer cell death in response to chemotherapy.

Just by looking at the cells chromatin structure, we could predict whether or not it would survive, says Backman. Cells with normal chromatin structures die because they cant respond; they cant explore their genome in search of resistance. They cant develop resistance.

Based on their discovery, the researchers hypothesized that altering the structure of chromatin to make it more orderly could be one way of boosting cancer cells vulnerability to treatment.

On further investigation, the team found that they could modify chromatins structure by altering electrolytes in the nucleus of cancer cells.

The team tested this strategy using two drugs that are already approved by the Food and Drug Administration (FDA): Celecoxib and Digoxin.

Celecoxib is currently used for pain relief, while Digoxin is used to treat atrial fibrillation and heart failure. Both drugs, however, are also able to change the packing density of chromatin.

The researchers combined these drugs which they refer to as chromatin protection therapeutics (CPTs) with chemotherapy and tested them on cancer cells in the laboratory. According to Backman, they witnessed something remarkable.

Within 2 or 3 days, nearly every single cancer cell died because they could not respond. The CPT compounds dont kill the cells; they restructure the chromatin. If you block the cells ability to evolve and to adapt, thats their Achilles heel.

Vadim Backman

While the researchers are excited by their findings, they caution that animal and human studies are needed before any firm conclusions can be made.

There is a big difference between cell cultures and humans, says Backman. You never know how the environment inside the human body will affect cancers behavior or if there will be unforeseen side effects.

That said, the researchers note that they have replicated their findings in seven different cancer types so far, which Backman says is very promising.

Visit link:
Cancer cells destroyed in just 3 days with new technique

Read More...

#ASH20: Orca’s team showcases their first cut of the data on promising cell purification work for HSCT – Endpoints News

Monday, December 7th, 2020

Ridding the world of Covid-19 is a lucrative enterprise.

As analysts divvy up the multibillion-dollar rewards in line for the leaders in the race to roll out the first pandemic vaccines, no one is forgetting to track just how much the top company founders have on the line now as stock prices gyrate ever higher.

Bloomberg, which tracks these numbers with missionary zeal, puts Sahins net worth today at $5.1 billion following the most recent rise of BioNTechs $BNTX share price. The biotechs partner, Pfizer, is aggressively pushing ahead with plans to get the first approved vaccine in play, and Bernstein believes that will put the two collaborators in line for the largest market share for next year.

Unlock this story instantly and join 94,800+ biopharma pros reading Endpoints daily and it's free.

SUBSCRIBE SIGN IN

Continue reading here:
#ASH20: Orca's team showcases their first cut of the data on promising cell purification work for HSCT - Endpoints News

Read More...

ASH 2020: Results from a Study of Inherited Blood Disorders Treated with CRISPR/Cas9 – OncoZine

Monday, December 7th, 2020

Results from a study using cutting-edge genome editing for hard-to-treat blood disorders were presented at the 62nd American Society of Hematology (ASH) Annual, Meeting and Exposition held virtually from December 5 8, 2020.

The researchers reported promising interim safety and efficacy data from 10 patients who received an investigational gene-editing based therapy, CTX001, the first to test a CRISPR-Cas9 gene-editing therapy in humans for a genetic disease.

Sickle cell disease affects approximately 100,000 people in the United States. And globally, approximately 400,000 infants are born each year with sickle cell disease. [3][4]

Recognized for its abnormally shaped red cells, sickle cell disease can cause a variety of health problems, including hemolytic anemia, episodes of severe pain, called vaso-occlusive crises, as well as progressive and irreversible organ damage, and strokes, leading to a decreased health-related quality of life (hrQoL), and early death.[5]

Standard of of careToday, the standard curative treatment for sickle cell disease is allogeneic hematopoietic stem-cell transplantation. If matched with a sibling donor, transplantation is known to be curative in more than 90% of patients. This approach is approach has limitations including a higher risk of complications in older patients, a risk of severe graft-versus-host disease (GVHD), and lack of an available matched sibling in approximately 80% of cases.[6][7]

Patients with transfusion-dependent thalassemia are dependent on blood transfusions from early childhood. The only available cure for both diseases is a bone marrow transplant from a closely related donor, an option that is not available for the vast majority of patients because of difficulty locating matched donors, the cost, and the risk of complications.

In the studies, the researchers goal is to functionally cure the blood disorders using CRISPR/Cas9 gene-editing by increasing the production of fetal hemoglobin, which produces normal, healthy red blood cells as opposed to the misshapen cells produced by faulty hemoglobin in the bodies of individuals with the disorders.

The clinical trials involve collecting stem cells from patients. Researchers edit the stem cells using CRISPR-Cas9 and infuse the gene-modified cells into the patients. Patients remain in the hospital for approximately one month following the infusion.

Prior to receiving their modified cells, the seven patients with beta-thalassemia required blood transfusions approximately every three to four weeks and the three patients with sickle cell disease suffered episodes of severe pain roughly every other month. All the individuals with beta-thalassemia have been transfusion independent since receiving the treatment, a period ranging between two and 18 months. Similarly, none of the individuals with SCD have experienced vaso-occlusive crises since CTX001 infusion. All patients showed a substantial and sustained increase in the production of fetal hemoglobin.

SafteyResearchers report that the safety of CTX001 infusion was generally consistent with the chemotherapy regimen received prior to cell infusion. Four serious adverse events related or possibly related to CTX001 were reported in one patient with thalassemia: headache, haemophagocytic lymphohistiocytosis (HLH), acute respiratory distress syndrome, and idiopathic pneumonia syndrome.

The patients have now recovered.

Unmet medical needThere is a great need to find new therapies for beta-thalassemia and sickle cell disease, said Haydar Frangoul, MD, Medical Director of Pediatric Hematology and Oncology at Sarah Cannon Research Institute, HCA Healthcares TriStar Centennial Medical Center.

What we have been able to do through this study is a tremendous achievement. By gene editing the patients own stem cells, we may have the potential to make this therapy an option for many patients facing these blood diseases, Frangoul concluded.

Because of the precise way CRISPR-Cas9 gene editing works, Frangoul suggested the technique could potentially cure or ameliorate a variety of diseases that have genetic origins.

Given that the only Food and Drug Administration (FDA) -approved cure for sickle cell disease, a bone marrow transplant, is not widely accessible, having another curative option would be life-changing for a large number of the sickle cell disease population, noted Catherine Bollard, MD, of Childrens National Research Institute and George Washington University.

While longer follow-up data are needed, this study is extremely exciting for the field, she added.

The trial was sponsored by CRISPR Therapeutics and Vertex Pharmaceuticals.

Abstract[1] Frangoul H, Bobruff Y, Cappellini MD, Corbacioglu S, Fernandez CM, De la Fuente J, Grupp SA, Handgretinger R, et al, Safety and Efficacy of CTX001 in Patients with Transfusion-Dependent - Thalassemia and Sickle Cell Disease: Early Results from the Climb THAL-111 and Climb SCD-121 Studies of Autologous CRISPR-CAS9Modified CD34+ Hematopoietic Stem and Progenitor Cells (Abstract #4)

Reference[2] Frangoul H, Altshuler D, Cappellini MD, Chen YS, Domm J, Eustace BK, Foell J, De la Fuente J, Grupp S, et al. CRISPR-Cas9 Gene Editing for Sickle Cell Disease and -ThalassemiaH. N Engl J Med. 2020 Dec 5; DOI: 10.1056/NEJMoa2031054[3] Heeney MM, Ware RE. Hydroxyurea for children with sickle cell disease. Hematol Oncol Clin North Am 2010;24:199-214.2.[4] Piel FB, Patil AP, Howes RE, et al. Global epidemiology of sickle haemoglobin in neonates: a contemporary geostatistical model-based map and population estimates. Lancet 2013; 381: 142-51[5] Esrick EB, Lehmann LE, Biffi A, Achebe M, Brendel C, Ciuculescu MF, Daley H, et al. Post-Transcriptional Genetic Silencing of BCL11A to Treat Sickle Cell Disease. N Engl J Med. 2020 Dec 5; DOI: 10.1056/NEJMoa2029392[6] Gluckman E, Cappelli B, Bernaudin F, et al. Sickle cell disease: an international survey of results of HLA-identical sibling hematopoietic stem cell transplantation. Blood 2017; 129: 1548-56.4.[7] Bernaudin F, Dalle J-H, Bories D, et al. Long-term event-free survival, chimerism and fertility outcomes in 234 patients with sickle-cell anemia younger than 30 years after myeloablative conditioning and matched-sibling transplantation in France. Haematologica 2020; 105: 91-101.

Featured image: The ASH Store at the American Society of Hematology 61th Annual Meeting at the Orange County Convention Center. Photo courtesy 2019. ASH/Scott Morgan. Used with permission.

Go here to see the original:
ASH 2020: Results from a Study of Inherited Blood Disorders Treated with CRISPR/Cas9 - OncoZine

Read More...

Poor COVID-19 Outcomes Among Patients With Hematologic Malignancies: Results From ECOVIDEHE – Cancer Therapy Advisor

Monday, December 7th, 2020

Hematologic malignancy was associated with more severe symptoms and death from coronavirus disease 2019 (COVID-19), likely due to immunosuppression attributable to the hematologic disease, according to results of a retrospective study presented at the virtual 62nd American Society of Hematology (ASH) Annual Meeting and Exposition.

The worse disease evolution should be taken into account for a population that is highly exposed to SARS-CoV-2 contagion due to [a] high number of hospital visits for treatment, Cristina De Ramn, of the Hospital Universitario de Salamanca in Spain, and presenter of the study, said.

Prior studies have suggested that hematologic disease is associated with higher COVID-19 mortality rates than the general population, but the data so far are limited. The aim of this study, called ECOVIDEHE, was to assess the effect of hematologic disease and its associated treatment with the outcomes of COVID-19.

The multicenter, retrospective observational study evaluated data from 543 patients with hematologic disease who developed COVID-19 between March and June 2020. Samples and data were collected at the time of assistance in the emergency department or hospital admission.

At baseline, the median age of patients was 70 years, 57% of patients were male, and 76% had at least 1 comorbidity. There were 65% of patients in the cohort with a lymphoid malignancy, of which, 53.6% were on active anticancer treatment. SARS-CoV-2 infection was confirmed with a positive nasopharyngeal swab or serologic testing among 94% of patients, and 15% were nosocomial.

There were 89% of patients who required hospital admission, including 6.3% who were admitted to intensive care units (ICU). Overall, 65% of patients were considered to have severe COVID-19. The most common symptoms were fever, cough, dyspnea, and pneumonia.

Overall, the mortality rate was 36.3%. Patients with acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS; 49%) were significantly more likely to die from COVID-19 than patients with other hematologic malignancies (34%) or other hemopathies (29%; P <.001). Patients with active or progressive disease were also more likely to die from COVID-19 (51%) compared with patients in complete remission (21%) or those with partial remission or stable disease (37%: P =.018).

Patients who had undergone an allogeneic (22%) or autologous hematopoietic stem cell transplant (20%) experienced improved survival compared with patients who had not undergone transplant (39%; P <.01). Ms De Ramn said that these findings may be because the patients who underwent transplant were younger and many were in complete remission.

In addition, mortality was associated with higher Charlson Comorbidity Index and Eastern Cooperative Oncology Group performance status. Mortality was also associated with low lymphocyte or platelet counts, as well as high lactate dehydrogenase, C-reactive protein, procalcitonin, and D-dimer levels. A multivariate analysis confirmed these findings, and also found that being older than 70 years was associated with mortality.

Most treatments for COVID-19 did not improve survival outcomes, except corticosteroids. Ms De Ramn suggested that this may be because the treatments were preferentially administered to patients with more severe disease.

Ms De Ramn concluded that SARS-CoV-2 infection causes more severe disease and higher mortality rates in hematological patients, especially those with AML/MDS or active or progressive disease.

Read more of Cancer Therapy Advisors coverage of the ASH 2020 meeting by visiting the conference page.

Reference

De Ramn C, Hernandez-Rivas JA, Garca JAR, et al. Impact of Sars-CoV2 infection on 491 hematological patients: the Ecovidehe Multicenter Study. Presented at: 62nd American Society of Hematology (ASH) Annual Meeting and Exposition; December 5-9, 2020. Abstract 312.

Read the original here:
Poor COVID-19 Outcomes Among Patients With Hematologic Malignancies: Results From ECOVIDEHE - Cancer Therapy Advisor

Read More...

Backed by a $200M investment from a Korean conglomerate, Vivek Ramaswamy has quietly built a ‘mother ship’ for vant creation with a whole new…

Monday, December 7th, 2020

Ridding the world of Covid-19 is a lucrative enterprise.

As analysts divvy up the multibillion-dollar rewards in line for the leaders in the race to roll out the first pandemic vaccines, no one is forgetting to track just how much the top company founders have on the line now as stock prices gyrate ever higher.

Bloomberg, which tracks these numbers with missionary zeal, puts Sahins net worth today at $5.1 billion following the most recent rise of BioNTechs $BNTX share price. The biotechs partner, Pfizer, is aggressively pushing ahead with plans to get the first approved vaccine in play, and Bernstein believes that will put the two collaborators in line for the largest market share for next year.

Unlock this story instantly and join 94,800+ biopharma pros reading Endpoints daily and it's free.

SUBSCRIBE SIGN IN

Read the original post:
Backed by a $200M investment from a Korean conglomerate, Vivek Ramaswamy has quietly built a 'mother ship' for vant creation with a whole new...

Read More...

ASH Goes Remote as CAR T-Cell Therapy Competition Heats Up – AJMC.com Managed Markets Network

Monday, December 7th, 2020

Updated data for a second anti-BCMA therapy, idecabtagene vicleucel (ide-cel) from Bristol Myers Squibb/bluebird bio, will be presented, including health-related quality of life results from the KarMMA study in patients with heavily pretreated R/R multiple myeloma. FDA has assigned a March 27, 2021, target date for action on this therapy.

Also anticipated are results from the APOLLO study in relapsed multiple myeloma, which will show that adding daratumumab and hyaluronidase-finj, called Darzalex Faspro by Janssen, to pomalidomide and dexamethasone reduces the risk of disease progression or death by 37% compared with pomalidomide and dexamethasone alone.

Notably, this phase 3 study involves subcutaneous administration of daratumumab, which offers significantly reduced treatment time and burden for patients. Janssen has submitted results from APOLLO to FDA and the European regulators.

The subcutaneous formulation of daratumumab offers patients and physicians a 3- to 5-minute administration experience and the potential to reduce systemic administration-related reactions compared to intravenous administration of daratumumab, said Meletios A. Dimopoulos, MD, professor and chairman of the Department of Clinical Therapeutics at the National and Kapodistrian University of Athens School of Medicine, Athens, Greece, who is the studys principal investigator.

The REACH3 study, a phase 3 randomized study of ruxolitinib (Jakavi) vs best-available-therapy, will have important implications in chronic graft-vs-host-disease (GvHD). This condition occurs when new T cells from a stem cell transplant identify the patients cells as foreign and attack them, creating reactions from rashes to gastrointestinal issues to harm to the liver.

Results involving transplant in myelodysplatic syndromes (MDS) could have important implications for reimbursement. Corey Cutler, MD, MPH, FRCPC, of Dana-Farber Cancer Institute will present results that show transplantation of hematopoietic stem cells from compatible donors nearly doubled the survival rate of patients aged 50 to 75 years.

Even though transplant is frequently used in younger patients, it has not been widely used among older patients. Lack of Medicare coverage is a major barrier, Cutler explained. This study adds to a growing body of evidence that suggests its time to revisit the reimbursement question.

Asked his thoughts on whether CMS might change its policy, Cutler said, I cant speak for the agency, but I will tell you there are several studies that do suggest it should be covered.We are, of course, reaching out to CMS.

Fridays press briefing ahead of the opening of ASH highlighted the results for MDS and daratumumab and others that are expected to be practice changing. To know that older patients do well with transplant is a really important message, said Lisa Hicks, MD, MSc, a hematologist from St. Michaels Hospital in Canada, who moderated the briefing.

Ian Flinn, MD, of Tennessee Oncology, who is an author on several studies being presented at ASH involving venetoclax (Venclexta) and Brutons tyrosine kinase (BTK) inhibitors in chronic lymphocytic leukemia, said he was interested to see the results of the CAPTIVATE trial.

He said that right now, venetoclax is a fixed-duration therapy. Now, we need to figure out whether thats a good idea or not, Flinn said. CAPTIVATE will help clinicians understand whether they should keep patients on venetoclax plus ibrutinib after they have reached the point of minimal residual disease.

The ASH meeting will also highlight research examining disparities in care, as well as the effects of COVID-19 on outcomes. On Saturday, Anthony Fauci, MD, director of the National Institute of Allergy and Infectious Diseases (NIAID), will discuss the latest information on COVID-19 and its impact on hematologic conditions in a fireside chat with ASH President Stephanie J. Lee, MD, MPH.

This week, President-elect Joe Biden announced that Fauci will be a chief medical adviser, in addition to retaining his longtime role at NIAID during the new administration.

Maggie L. Shaw contributed to this report.

See the original post here:
ASH Goes Remote as CAR T-Cell Therapy Competition Heats Up - AJMC.com Managed Markets Network

Read More...

Gene therapy gives man with sickle cell disease the chance for a better future – UCLA Newsroom

Thursday, December 3rd, 2020

For Evie Junior, living with sickle cell disease has been like running a marathon.

But its a marathon where as you keep going, the trail gets rockier and then you lose your shoes, the 27-year-old said. It gets harder as you get older. Things start to fail and all you can think about is how much worse its going to get down the road.

In sickle cell disease, a genetic mutation causes the blood-forming stem cells which give rise to all blood and immune cells to produce hard, sickle-shaped red blood cells.These misshapen cells die early, leaving an insufficient number of red blood cells to carry oxygen throughout the body. Because of their sickle shape, these cells also get stuck in blood vessels, blocking blood flow and resulting in excruciating bouts of pain that come on with no warning and can leave patients hospitalized for days.

The disease affects 100,000 people in the United States and millions around the world, the majority of whom are of African or Hispanic descent. It can ultimatelylead to strokes, organ damage and early death.

As a child growing up in the Bronx, New York, Junior had to have his gall bladder and spleen removed due to complications from the disease, but he refused to let his condition limit him. He played football, basketball and baseball during the day, even though on some nights he experienced pain crises so severe he couldnt walk.

It was just really routine if I had a sickle cell crisis, he said. Going to the emergency room, staying in the hospital, coming out in a few days and then getting back to normal life.

I want to create a better future

When he was 24 and living in Portland, Oregon, Junior began working as an emergency medical technician. He adopted the same mentality trying to treat his pain episodes the best he could, and hoping they would resolve overnight so he could get back to work. Around that time, though, the crises became harder to manage. He developed pericarditis, an inflammation in the layers of tissue around his heart, and needed six weeks to recover.

The big worry with sickle cell disease is that youre going to die young from some type of complications or damage to your organs, he said. In the last couple of years, Ive been seeing that slowly happen to me and I can only suspect that its going to keep getting worse. I want to create a better future for myself.

In July 2019, in pursuit of that future, Junior enrolled in a clinical trial foran experimental stem cell gene therapy for sickle cell disease. The study is led byUCLA Broad Stem Cell Research Centerphysician-scientistsDr. Donald KohnandDr. Gary Schillerand funded by the California Institute for Regenerative Medicine.

The therapy, developed by Kohn over the past 10 years, is intended to correct the mutation in patients blood-forming stem cells to allow them to produce healthy red blood cells.Kohn has already applied the same concept to successfully treat several immune system deficiencies, includinga cure for a form of severe combined immune deficiency, also known as bubble baby disease.

But sickle cell disease has proven more difficult to treat with gene therapy than those other conditions. Junior volunteered for the trial knowing there was a chance the therapy wouldnt cure him.

Even if it doesnt work for me, Im hoping that it can be a cure later down the road for millions of people, he said.

In July 2020, Junior received an infusion of his own blood-forming stem cells that had been genetically modified to overcome the mutation that causes his disease.

The goal of this treatment is to give him a future, let him plan for college, family or whatever he wants without worrying about getting hospitalized because of another pain crisis, said Kohn,a distinguished professor of microbiology, immunology and molecular genetics, pediatrics, and molecular and medical pharmacology at theDavid Geffen School of Medicine at UCLA.

Reason for optimism

Three months after his treatment, blood tests indicated that 70% of Juniors blood stem cells had the new corrected gene. Kohn and Schiller estimate that even a 20% correction would be enough to prevent future sickle cell complications. Junior said he hasnt had a pain crisis since undergoing the treatment and he has more energy and feels out of breath less often.

I noticed a big difference in my cardiovascular endurance in general even going for a light jog with my dogs, I could feel it, he said.

Junior and his doctors are cautiously optimistic about the results.

Its too early to declare victory, but its looking quite promising at this point, Kohn said. Once were at six months to a year, if it looks like it does now, Ill feel very comfortable that hes likely to have a permanent benefit.

After a lifetime of dealing with the unwelcome surprises of the disease, Junior is even more cautious than his doctors. But as the weeks pass, hes slowly allowing a glimmer of hope that he could soon be someone who used to have sickle cell disease. For him, that hope feels like a burst of happiness thats followed by thoughts of all the things he could do with a healthy future: pursue his dream of becoming a firefighter, get married and start a family.

I want to be present in my kids lives, so Ive always said Im not going to have kids unless I can get this cured, he said. But if this works, it means I could start a family one day.

Here is the original post:
Gene therapy gives man with sickle cell disease the chance for a better future - UCLA Newsroom

Read More...

Stem cell therapy in coronavirus disease 2019: current evidence and future potential – DocWire News

Thursday, December 3rd, 2020

This article was originally published here

Cytotherapy. 2020 Nov 9:S1465-3249(20)30932-4. doi: 10.1016/j.jcyt.2020.11.001. Online ahead of print.

ABSTRACT

The end of 2019 saw the beginning of the coronavirus disease 2019 (COVID-19) pandemic that soared in 2020, affecting 215 countries worldwide, with no signs of abating. In an effort to contain the spread of the disease and treat the infected, researchers are racing against several odds to find an effective solution. The unavailability of timely and affordable or definitive treatment has caused significant morbidity and mortality. Acute respiratory distress syndrome (ARDS) caused by an unregulated host inflammatory response toward the viral infection, followed by multi-organ dysfunction or failure, is one of the primary causes of death in severe cases of COVID-19 infection. Currently, empirical management of respiratory and hematological manifestations along with anti-viral agents is being used to treat the infection. The quest is on for both a vaccine and a more definitive management protocol to curtail the spread. Researchers and clinicians are also exploring the possibility of using cell therapy for severe cases of COVID-19 with ARDS. Mesenchymal stromal cells are known to have immunomodulatory properties and have previously been used to treat viral infections. This review explores the potential of mesenchymal stromal cells as cell therapy for ARDS.

PMID:33257213 | DOI:10.1016/j.jcyt.2020.11.001

Read the original:
Stem cell therapy in coronavirus disease 2019: current evidence and future potential - DocWire News

Read More...

HCT Deemed More Favorable for MDS That Is High Risk – Cancer Therapy Advisor

Thursday, December 3rd, 2020

Hematopoietic stem cell transplant (HCT) treatment of myelodysplastic syndromes (MDS) resulted in a lower risk of death if performed early after diagnosis or among patients with high-risk disease, according to results of an observational study published in Leukemia.

HCT is the only potentially curative treatment for MDS. However, the disease primarily occurs among older patients in whom myoablative conditioning regimens are associated with an increased risk of mortality. The aim of this study was to evaluate mortality after transplant in a real-world population of patients with MDS.

The prospective, observational study included 290 patients with advanced MDS aged 60 to 75 years from the MDS Transplant-Associated Outcomes study who were determined to be fit to undergo HCT. All patients had disease that warranted consideration of HCT, including patients with intermediate- to high-risk disease or those with standard risk and severe cytopenia. The primary endpoint was overall survival (OS).

At baseline, the median age was 69 years, and the majority of patients had intermediate-risk disease (78%) and good cytogenetic risk (53%). Mutations in TP53, JAK2, or in the RAS pathway were either not present or unknown in most patients.

HCT was performed in 113 patients (39%) with a median time to HCT of 5 months (range, 1-58 months).

The median OS of the entire cohort was 29 months (95% CI, 19-39.5 months), which translated to a 3-year OS of 46%. Overall, there was no difference in the risk of death between the cohort who underwent HCT and those who did not (hazard ratio [HR], 0.75; 95% CI, 0.52-1.09; P =.13), in a multivariate analysis. However, landmark analyses showed a survival benefit with HCT at 5 (P =.04) and 9 months (P =.01), but not at 12 months or 24 months.

HCT performed earlier, within 5 months from study entry, was significantly associated with a lower risk of death than transplants performed later (HR, 0.53; 95% CI, 0.33-0.83; P =.006). HCTs performed after 5 months did not result in a survival benefit between the HCT and non-HCT cohorts.

Transplant also lowered the risk of death among patients with adverse-risk disease compared with patients with standard-risk MDS with severe cytopenia (HR, 0.57; 95% CI, 0.37-0.88; P =.01).

Other characteristics associated with a survival benefit with HCT included patients with poor cytogenetic risk, an Eastern Cooperative Oncology Group performance status of 1, and patients of male sex.

The authors concluded that these data showed that there were also significant benefits for those receiving HCT within 5 months and for those with adverse disease risk factors as compared to standard risk with severe cytopenias.

Reference

Abel GA, Kim HT, Hantel A, et al. Fit older adults with advanced myelodysplastic syndromes: who is most likely to benefit from transplant? Leukemia. Published November 17, 2020. doi:10.1038/s41375-020-01092-2

Read the original:
HCT Deemed More Favorable for MDS That Is High Risk - Cancer Therapy Advisor

Read More...

Not All Patients With Relapsed DLBCL Referred for CAR T in Community Setting – Targeted Oncology

Thursday, December 3rd, 2020

Hematologists and oncologists working in the community setting encounter multiple obstacles when prescribing chimeric antigen receptor (CAR) T-cell therapy to patients with relapsed or refractory diffuse large B-cell lymphoma (DLBCL). The challenges involve matters of processes, treatment cost, and access to treatment.

To further understand the issues and the solutions needed for physicians who treat relapsed/refractory DLBCL, researchers at Cardinal Health conducted 2 live survey sessions to collect information from clinicians. A total of 114 oncologists and hematologists from community practices and hospital settings participated in the survey. The population of hematologists/oncologists see roughly 20 patients per day, and the majority have been in practice for 11 to 20 years. Overall, 46% of the clinicians who attended the first live survey session, and 26% of those who attended the second reported that they had not referrer any patient for CAR T-cell therapy, and of those who did refer patients 32% and 22% of patients, respectively had not yet been infused with CAR T cells.1

The results of the survey revealed that while the use of CAR T-cell therapy increased in community practices over the past year, there remain issues with high cost and toxicity of treatment. It was also reported that the processing of insurance was a barrier to getting patients treated. These challenges continue to limit the number of clinicians who recommend CAR T-cell therapy to their patients.

In an interview withTargeted Oncology, Ajeet Gajra, MD, FACP, vice president, Cardinal Health, discussed the ongoing challenges community oncologists face with prescribing CAR T-cell therapy to patients with relapsed/refractory DLBCL.

TARGETED ONCOLOGY: Can you explain the overall prognosis for patients with DLBCL? What are outcomes generally like with existing standard of care therapy?

Gajra: The outlook for DLBCL improved with the advent of chemoimmunotherapy, better risk stratification, and improved supportive care. Recent studies demonstrate that despite aggressive biology, over 60% of patients with DLBCL treated with chemoimmunotherapy achieve long-term remissions and cures. However, the improvements reached a plateau in the past decade, especially for patients who relapse after initial chemoimmunotherapy. These patients typically have poor prognostic features as defined by the International Prognostic Index (IPI) with high likelihood of relapse and death. Patients with relapsed or refractory disease are typically treated with salvage immunochemotherapy such as rituximab, ifosfamide, carboplatin and etoposide (RICE) or rituximab, cisplatin high dose Ara-C and dexamethasone (RDHAP), and those with chemotherapy-sensitive disease receive autologous stem cell transplant (ASCT). Using this approach, complete response (CR) rates are 35% to 40%, and in a recent study the 3-year event-free survival (EFS) and overall survival (OS) were 31% and 50%, respectively. Outcomes with ASCT are much worse for patients with refractory DLBCL as demonstrated in the SCHOLAR trial wherein the objective response rate was 26% (CR rate, 7%) with a median OS of 6.3 months and only 20% of patients were alive at 2 years.

Thus, prior to 2017 when the first CAR T therapy was approved in DLBCL with progression after 2 prior lines of therapy, there had been a significant unmet need for patients with relapsed DLBCL. The approval of 2 CAR-T therapies, axicabtagene ciloleucel (axi-cel) in October of 2017 and tisagenlecleucel (Kymriah; tisa-cel) in May 2018, in the treatment of large-cell lymphoma (LBCL), has ushered in a new mode of treatment which offers the potential of long-term remission in what was essentially a fatal disease.

TARGETED ONCOLOGY: What has been your observation experience with using CAR T cell therapy in patients with DLBCL by US community oncologists?

Gajra: Axi-cel and tisa-cel are both CD19-directed, genetically modified autologous T cell immunotherapy agents. Since the process of obtaining CAR T therapy for an individual patient is quite complex, we sought to assess the uptake of these agents among United States community oncologists. We conducted a study of community oncologists at two time points to assess perceptions and use of approved CAR T therapies in relapsed DLBCL. At each time point over 50 distinct oncologists participated. At the early timepoint, 46% of participants indicated that they had not referred any patients for CAR T therapy but at the later timepoint, this number decreased to 29% suggesting increasing use over the course of the 10-month interval. Of those participants who had referred patients for CAR T therapy, 32% at the early timepoint reported that none of their patients had yet received the CAR T infusion but the percentage of non-receipt decreased to 22% at the later timepoint again suggesting improved uptake and utilization.

TARGETED ONCLOGY: How do patient characteristics factor into how oncologists select patients to administer CAR T cells to? What are the barriers to CAR-T use?

Gajra: CAR T therapies approved in DLBCL have limitations as defined by the FDA approval and are to be used in adult patients with relapsed or refractory large B-cell lymphoma, including DLBCL, after 2 or more lines of systemic therapy. Neither agent is approved for the use of CNS lymphoma. As with the pivotal trials for the 2 agents, patients must have good ECOG performance status, adequate organ function including marrow, hepatic, cardiac and renal function, no active infection and no CNS involvement. Both agents carry black box warnings for neurotoxicity and cytokine release syndrome (CRS) which can be potentially fatal. Thus, the patients selected need to have good physiologic reserve and be willing to accept risks associated with the therapies. With the approval of a new CD19-directed monoclonal antibody, tafasitamab, it is not clear if patients exposed to that agent can still benefit from CAR T therapies.

In addition to patient specific factors, CAR T therapy represents a complex manufacturing process that is unlike traditional drug therapy or stem cell transplant. After identification of a potential patient with relapsed LBCL who has received at least two prior systemic therapies, a benefits verification and referral to a designated CAR T-cell therapy center is required. If deemed appropriate by the CAR T center, the patient undergoes apheresis for T-cell collection. The cells are then transported to the manufacturers facility where they are isolated, activated and undergo gene transfer, creating the chimeric cells which go through a process of expansion to generate the numbers needed for therapeutic effect. This process takes from 10 days to a few weeks. The CAR T cells are then cryopreserved and transferred back to the CAR T facility and reinfused into the patient. Thus, it is critical to maintain vein to vein integrity. Thus, unlike traditional cytotoxic or monoclonal antibody products, these agents are patient specific, living cell products that have a complex process for their manufacture, storage and shipping, leading to high costs to the healthcare system and the patient.

Given this information, not surprisingly, the oncologists surveyed identified the high cost of therapy as a major barrier to uptake and utilization at both time points respectively. Over half the participants identified cumbersome logistics of administering therapy and following patients as another major barrier. Further exploration of logistical issues identified barriers encountered during the referral process could be attributed to the payer or the CAR T center.

The payer specific challenges identified include slow approval process by 27% of payers (and high rates of denials by in 13% of payers. The challenges specific to the CAR-T center include slow intake process by 23% of CAR T centers lack of a CAR T center in geographic vicinity in 13%. CAR T center choosing stem cell transplant rather than CAR T for the patient was also seen 10% of the time. Other commonly encountered clinical challenges reported by the participants included deterioration of the patient prior to CAR T administration, and the need to administer bridging chemotherapy while awaiting manufacture of CAR T therapy. The lack of communication from the CAR T center during the process was identified by a minority as an impediment to recommending CAR T therapies, including lack of instructions to the primary oncologist and the patient.

TARGETED ONCOLOGY: Can you discuss the toxicities observed with CAR T cell therapy in this patient population? Do you haveany insight into toxicities observed in the real-world setting?

Gajra: As stated, both approved products carry black box warnings for CRS and neurotoxicity, now called Immune Effector Cell Associated Neurologic Syndrome (ICANS). CRS is an acute systemic inflammatory syndrome characterized by fever, hypotension, tachycardia, hypoxia and multiple organ dysfunction. ICANS is a neuropsychiatric complex manifested by encephalopathy, headache, tremor, dizziness, aphasia, delirium, insomnia and anxiety. The treating team needs to maintain a high index of suspicion for these potentially life-threatening agents and patients need to have access to facilities with advanced critical care. Tumor debulking ahead of CAR T infusion and prophylactic use of tocilizumab may reduce the risk of CRS. Use of corticosteroids early can alleviate the severity and duration of ICANS.

The scientific team at Cardinal Health has studied the real-world adverse events (AEs) to CAR T agents in DLBCL.2 We analyzed the postmarketing case reports from the FDA, AEs reporting system involving axicel and tisa-cel for large B-cell lymphomas were analyzed. Of 804 AE cases identified 67% of axi-cel cases and 26% of tisa-cel cases reported neurological AEs. Compared with cases without neurological AEs, significant associations were observed between neurological AEs and use of axi-cel, age 65 years, CRS and the outcome of hospitalization. These findings and those of other investigators suggest that there may be differences in neurological toxicity based on the agent used.

TARGETED ONCOLOGY: Can you provide background on how this web-based survey can about at Cardinal Health Specialty Solutions? What is the overall goal with it?

Gajra: We are continuously engaged in research with healthcare providers, including medical oncologists/hematologists, to assess their perspectives on issues they face in their day-to-day practice, including the impact of new therapies on patient care. We share our research findings with healthcare stakeholders through peer-reviewed manuscripts and abstracts, as well as through our Oncology Insights report, which is published twice a year.

TARGETED ONCOLOGY:How can the information obtained from this survey impact practice? Where are you in the process of response collect and obtaining results?

Gajra: Our research on CAR-T therapy, collected via web-based and in-person surveys, has helped us identify the challenges to the use of these therapies encountered by community oncologists. Given that over 50% of cancer care is rendered in the community setting, it is important to identify these barriers with a goal of mitigating them and facilitating timely access to these potentially life-saving therapies for patients. With a new CAR-T approval in mantle cell lymphoma this year and other potential approvals in newer indications on the horizon, streamlining access to CAR-T therapies will continue to be a priority.

We have a follow-up to this paper that will be presented at ASH 2020 where additional research with community oncologists in early 2020 has revealed that the rate of non-receipt of CAR-T therapies in DLBCL is relatively constant at around 30%. In addition, we are exploring interest and uptake of CAR-T therapies in the outpatient setting as oncologists gain more confidence in preventing, minimizing and managing the toxicity of CAR-T therapies.

References:

1. Gajra A, Jeune-Smith Y, Yeh T, et al. Perceptions of community hematologists/oncologists on barriers to chimeric antigen receptor T-celltherapy for the treatment of diffuse large B-cell lymphoma. Immunotherapy. 202012(10);725-732. doi: 10.2217/imt-2020-0118

2. Gajra A, Zettler ME, Phillips EG Jr, Klink AJ, Jonathan K Kish, Fortier S, Mehta S, Feinberg BA. Neurological adverse events following CAR T-cell therapy: a real-world analysis. Immunotherapy. 2020 Oct;12(14):1077-1082. doi: 10.2217/imt-2020-0161

Read more:
Not All Patients With Relapsed DLBCL Referred for CAR T in Community Setting - Targeted Oncology

Read More...

Turkey’s Covid death toll hits record for ninth day as it happened – The Guardian

Thursday, December 3rd, 2020

New Covid-19 infections in France stayed below 10,000 for the third day in a row on Tuesday, a sequence unseen since mid-September, and the number of people hospitalised resumed a downward trend.

The government has loosened its second national coronavirus lockdown, put in place on 30 October, by allowing all shops to reopen at the weekend.

The French president Emmanuel Macron said last week the lockdown could be lifted on 15 December if by then the number of new cases per day fell to 5,000 and the number of patients in intensive care declined to between 2,500 and 3,000.

Earlier in the day, Macron said that France should be in a position to embark on a broader Covid-19 vaccination campaign between April and June next year, after initially targeting a smaller group of people.

Health authorities reported 8,083 new cases over the past 24 hours on Tuesday, versus 4,005 on Monday and 9,784 on Sunday.

The seven-day moving average of daily new infections fell below 11,000 for the first time since 2 October, at 10,965, four times lower than the all-time high of 54,440 reached on 7 November.

The cumulative number of Covid-19 cases now totals 2,230,571, the fifth-highest in the world.

After increasing by 90 between Sunday and Monday, the number of people hospitalised for the disease fell by 619 to 27,639, below 28,000 for the first time since 4 November. The number of patients in ICUs declined by 146 to 3,605.

The number of people in France who have died from Covid-19 rose by 775 to 53,506, versus a rise of 406 on Monday. But the seven-day moving average of daily additional deaths stood at 467, below 500 for the first time in more than three weeks.

Read more here:
Turkey's Covid death toll hits record for ninth day as it happened - The Guardian

Read More...

Penn researchers find a way through the labyrinth keeping CAR-T from solid tumors – Endpoints News

Thursday, December 3rd, 2020

Blood vessels are supposed to act like trees, pumping in oxygen tissues need to grow and immune cells required to clear out pathogens. But in tumors, the forest can go a bit haywire. Vessels grow prodigiously and bulge and twist at abrupt points, making it difficult to even tell whats a vein and whats an artery. It starts to look less like a forest and more like a gnarled root floor. A disorganized labyrinth, one oncologist has called it.

For cancer, chaos is a virtue. That gnarled root floor insulates solid tumors from immune cells and, in recent years, has flustered drug developers best attempts at developing therapies meant to rev up the immune system and direct it toward the tumors.

Researchers at the University of Pennsylvania, however, think they may have stumbled onto a solution, a way of whipping the blood vessels back into proper shape. If it works, experts say, it could pave the way for CAR-T treatments that attack solid tumors and potentially improve the effectiveness for more traditional approaches, such as radiation and chemotherapy.

Its a really novel and potentially important approach, Patrick Wen, a neuro-oncologist at Dana-Farber who was not involved in the work, told Endpoints News. They really did good work. This is a very different way of improving immunotherapy.

Yi Fan, a radiation oncologist at Penns School of Medicine, has been working for the last few years to understand why the labyrinth appears in the first place. Researchers had previously circled in on the so-called growth factors that stimulate blood vessel formation. Attempts to block these factors, though, disappointed; Avastin, an antibody against the factor VEGF, became a blockbuster but has continually failed to improve survival on a range of malignancies.

Scientists would have to go more fundamental. In a pair of 2018 papers, Fan showed that part of the problem is a process called endothelial cell transformation. Cells lining the blood vessels around the tumor acquire stem cell-like properties that allow them to reproduce and expand rapidly, as stem cells do.

Theres a genetic reprogramming, Fan told Endpoints. Theyll become really aggressive.

But how did that reprogramming happen? If Fan could pin down the pathway, he figured he could then devise a way to block it. He started knocking out kinases the cellular engines that can drive epigenetic change, or reprogramming one by one in endothelial cells isolated from patients with an aggressive brain cancer called glioblastoma. Out of 518, 35 prevented transformation and one did so particularly well: PAK4.

Then they injected tumors into mice, some who had PAK4 and some who had the kinase genetically removed: Eighty percent of the mice who had PAK4 removed lived for 60 days, while all of the wild-type mice died within 40. Fans team also showed that T cells infiltrated the tumors more easily in the PAK4-less mice.

It was a fortuitous finding: Drug companies had developed several PAK inhibitors a decade ago, when kinase inhibitors were the flashiest thing in pharma. Many had been abandoned, but Karyopharm had recently brought a PAK4 blocker into Phase I.

To see whether drug developers could exploit this finding, Fan and his team removed T cells from mice and developed a CAR-T therapy to attack the tumors.

They gave mice three different regimens. The CAR-T therapy on its own failed to reduce tumor size, apparently unable to reach through the vessels. The Karyopharm drug also had little effect on its own. But combined, they managed to reduce tumor size by 80% after five days. They published the results in Nature Cancer this week.

It is a really eye-opening result, Fan said. I think we see something really dramatic.

That, of course, is just in mice, but Fan already has strong supporting evidence for PAK4s role in cancer. Last December, while Fan was still completing his experiment, Nature Cancer published a paper from Antoni Ribas UCLA lab suggesting that PAK4 inhibitors can help T cells infiltrate around various solid tumors. They showed that the same Karyopharm inhibitor could boost the effects of PD-1 inhibitors in mice, allowing activated T cells to better reach tumors.

That work has already translated into the clinic; weeks after it came out, Karyopharm added an arm to their Phase I study of the drug that will look at the PAK4 inhibitor in combination with the PD-1 blocker Opdivo.

Ribas said that Fans work is compelling and helps confirm the role of PAK4, but he said a CAR-T therapy would face a much longer path to the clinic. Its simply much easier to combine an approved drug with an experimental one than to devise a new CAR-T therapy, mix it with the unapproved inhibitor (and all the other things, such as bone marrow-clearing chemotherapy, CAR-T recipients receive) and then deduce what effect each is having.

It will a take a while, Ribas told Endpoints. But I hope this is right and its developed clinically.

There are also other unresolved obstacles for CAR-T in solid tumors, Wen said. Developers still struggle to find targets that wont also send the super-charged T cells after healthy tissue. And tangled blood vessels are just one of several mechanisms tumors have of defending themselves. They can, for example, turn tumor-eating immune cells into tumor-defending ones.

Still, Wen said, in the short term, the approach offered a path toward boosting the efficacy of radiation, chemotherapy and other small molecule drugs. Although Fan focused on glioblastoma, researchers agreed PAK4 likely plays the same vessel-warping role in many other solid tumors.

Theres a lot of things you could look at, he said.

In a January review, Jessica Fessler and Thomas Gajewski at the University of Chicago said Ribas paper pointed towards a path for improving PD-1 and overcoming resistance in some tumors. But they also raised questions about the Karyopharm drug, noting that it hits other proteins besides PAK4. That could mean other mechanisms are also at play and that the drug could affect other tissues in humans.

Ribas agreed that Karyopharms drug might not be the perfect molecule but said others could be on their way. He serves as a scientific advisor to Arcus, the Terry Rosen startup that is now working on developing its own PAK4 inhibitor.

If they can develop a very selective PAK4 inhibitor, he said, it may be a more direct way of testing the role of PAK4.

Tests with that drug, in turn, could help clear up a biological mystery that emerged out of Fans and Ribas papers. Although both investigators zeroed in on PAK4, each of them suggested very different mechanisms by which PAK4 kept immune cells out of the tumor. Ribas suggested it directly suppresses T cells, while Fan found it led to those transformations inside the blood vessels near the tumor.

Kinases are versatile proteins and both researchers said its possible that PAK4 is doing both. Its also possible, they said, that one is more important than the other, or simply that one of them is just wrong.

When you start with completely new biology, its hard to get it right the first time, Ribas said.

Original post:
Penn researchers find a way through the labyrinth keeping CAR-T from solid tumors - Endpoints News

Read More...

Nobel Prize history from the year you were born – Auburn Citizen

Thursday, December 3rd, 2020

Since 1901, Nobel Prizes have honored the worlds best and brightest and showcased the work of brilliant and creative minds, thanks to Swedish businessman Alfred Nobel, who made his fortune with the invention of dynamite.

The Prize in Physiology or Medicine often honors those whose discoveries led to medical breakthroughs, new drug treatments, or a better understanding of the human body that benefit us all.

The Prize in Literature celebrates those skilled in telling stories, creating poetry, and translating the human experience into words. The Prizes in Chemistry and Physics remind most of us how little we understand of genetics, atomic structures, or the universe around us, celebrating the scientists who further knowledge. A later addition to the award roster, the Nobel Memorial Prize in Economic Sciences is not an original Prize, but was established by the Central Bank of Sweden in 1968 as a memorial to Alfred Nobel. It applauds those who can unravel the mysteries of markets, trade, and money.

The Peace Prize celebrates, in Nobels words, the person who shall have done the most or the best work for fraternity between nations, the abolition or reduction of standing armies and for the holding and promotion of peace congresses, sometimes risking their lives to do so.

So precious are the awards that the medals of German physicists Max von Laue and James Franck, stored away for safekeeping in Copenhagen during World War II, were dissolved in acid to keep them away from approaching Nazi troops. After the war, the gold was reconstituted from the acid and recast into new medals.

But Nobel history has not been entirely noble. In 1939, British Prime Minister Neville Chamberlain, known for his policy of appeasement toward Nazi Germany, was nominated for the Peace Prize. In an act of irony and protest, members of the Swedish Parliament nominated Adolf Hitler. That nomination was withdrawn. Some recipients have ordered oppressive crackdowns on their own people or ignored genocides, either before or after receiving the Prize. The 1918 Nobel Prize in Chemistry was given to Germanys Fritz Haber, who invented a method of producing ammonia on a large scale, which was helpful in making fertilizer. But the same chemist helped develop the chlorine gas that was used as a chemical weapon in World War I.

Stacker looked at facts and events related to the Nobel Prizes each year from 1931 to 2020, drawing from the Nobel Committees recollections and announcements, news stories, and historical accounts.

Take a look, and see what was happening with the Nobel Prizes the year you were born.

You may also like: 100 years of military history

View original post here:
Nobel Prize history from the year you were born - Auburn Citizen

Read More...

PBRM1 and the glycosylphosphatidylinositol biosynthetic pathway promote tumor killing mediated by MHC-unrestricted cytotoxic lymphocytes – Science…

Thursday, December 3rd, 2020

INTRODUCTION

Cytotoxic lymphocytes (CLs) are subtypes of immune cells capable of detecting and killing tumor and virus-infected cells (1, 2). Current cancer immunotherapy mainly harnesses the antitumor activities of cytotoxic T cells, effectors of the adaptive immune system (25). T cells detect transformed cells by using their T cell receptors to recognize tumor neoantigens displayed on the major histocompatibility complex (MHC) class I proteins of tumor cells (68). T cellbased therapies such as checkpoint inhibitors substantially extend the survival of a subset of patients with cancer (35, 9). Many tumors, however, are unresponsive to checkpoint therapies, possibly because of defects in presentation of tumor antigens on MHC class I (2). Moreover, tumor cells that initially respond to checkpoint inhibitor therapies often develop acquired resistance, resulting in tumor relapses (2, 4). Therefore, additional immunotherapeutic strategies are needed to achieve sustained clinical benefits in a larger patient population.

Another class of tumor-killing CLs are MHC-unrestricted CLs, which include natural killer (NK) cells, effectors of the innate immune system, and NK-like cell lines (1014). Unlike T cells, MHC-unrestricted CLs kill tumor or virally infected cells without requiring prior activation by MHC-presented antigens (6, 15, 16). The absence of MHC class I on target cells serves as a strong activation signal for MHC-unrestricted CLs (1, 6, 10, 12, 17). As a result, MHC-unrestricted CLs are capable of detecting and destroying tumor cells resistant to T cellmediated attack, holding great promise as mono or combination immunotherapies (18, 19). However, the antitumor activities of MHC-unrestricted CLs have not been translated into significant clinical benefits to patients, likely because of escape mechanisms evolved by tumor cells to circumvent killing (19, 20). To improve the efficacy of MHC-unrestricted CLs in immunotherapy, it is critical to identify genes controlling the response of tumor cells to cytotoxicity.

In this work, we performed genome-wide genetic screens to uncover tumor-intrinsic genes that regulate tumor killing by MHC-unrestricted CLs. Our screens identified a large number of regulators that either promote or inhibit the response of tumor cells to killing. The screens isolated known mediators of NK cellmediated killing, but most of the identified genes were not previously linked to tumor killing. Multiple top-ranking genes in the screens belong to the glycosylphosphatidylinositol (GPI) anchor biosynthetic pathway, which is not involved in T cellmediated killing. Further analyses revealed that the GPI biosynthetic pathway is required for the activation and cytolytic granule secretion of CLs. Another critical regulator identified in the screens was PBRM1, a component of the SWI/SNF chromatin-remodeling complex. Notably, PBRM1 promotes the killing of tumor cells by MHC-unrestricted CLs, in contrast to its inhibitory activity in T cellmediated killing. Thus, PBRM1 plays opposite roles in MHC-restricted and MHC-unrestricted cytotoxicity. Like the GPI biosynthetic pathway, PBRM1 promotes cytolytic granule secretion in CLs. The factors identified in this work could represent potential targets for cancer immunotherapy.

We developed a tumor-killing platform using TALL-104 cells, a clinically relevant CL cell line exhibiting robust MHC-unrestricted cytotoxicity against human tumors (14, 18, 2123). Despite the T cell origin of this cell line, TALL-104 cells express NK cell receptors and closely resemble NK cells in recognizing and destroying tumors without prior sensitization (fig. S1) (14). Previously, TALL-104 cells have been used in clinical trials to treat patients with cancer (14, 18, 2124). A major advantage of TALL-104 cells is that they can be readily expanded into large homogeneous populations to meet the demands of genome-wide genetic screens. We observed that TALL-104 cells efficiently killed HAP1 cells (Fig. 1A and fig. S2A), a haploid human cell line derived from chronic myeloid leukemia cells (2527). By contrast, primary human cells, including peripheral blood mononuclear cells (PBMCs) and adipose stem cells (ASCs), were resistant to TALL-104mediated killing (Fig. 1A and fig. S2B), consistent with the ability of CLs to distinguish between tumor and normal cells (68).

(A) TALL-104 cells (CLs) kill HAP1 cells (tumor cells) but not normal cells. HAP1 cells and human PBMCs grown on a 24-well plate were treated with the indicated ratios of TALL-104 cells for 8 hours. The cells were stained with propidium iodide (PI) and measured by flow cytometry. Error bars indicate SD (n = 3). P values were calculated using Students t test. ***P < 0.001. n.s., P > 0.05. (B) Illustration of the genome-wide haploid genetic screen aiming to identify tumor-intrinsic genes required for TALL-104 cytotoxicity. (C) Bubble graph showing significant hits from the haploid genetic screen. The y axis depicts the log10 of P values for the gene hits in the TALL-104selected population as compared to a published unselected control (62) using Fishers exact test. Dashed line indicates the cutoff of significance. We set a P value cutoff of 1 105 to account for multiple hypothesis testing. In addition, for genes with P values less than 1 1010, we considered a gene as a hit only if it also had strong enrichment for sense-strand intron insertions based on the binomial test (P value cutoff: 1 105). The x axis depicts the chromosomal positions of the genes. The size of a circle is scaled according to the number of unique inactivating gene-trap insertions within the gene. Circles are colored according to the annotated or predicted functions of the gene products. Genes that did not reach the cutoff are shown in gray. Full datasets of the screen are included in table S1.

To systematically identify tumor-intrinsic genes required for TALL-104mediated killing, pooled HAP1 cells were randomly mutagenized using retroviral gene-trap insertions (2729). The mutagenized HAP1 cells were incubated with TALL-104 cells for four consecutive rounds, in which approximately 50% of the HAP1 cells were killed during each round (Fig. 1B). After the final round of killing, the TALL-104resistant HAP1 population was harvested, and their gene-trap retroviral insertions were mapped by deep sequencing. The gene-trap insertions were then compared to those of an untreated control HAP1 population to identify significant hits on the basis of the enrichment of inactivating gene-trap insertions.

The haploid genetic screen identified a number of genes that promote TALL-104mediated killing (Fig. 1C and table S1). Mutations of these genes in target cells caused resistance to TALL-104mediated cytotoxicity. One gene identified in the screen was NCR3LG1/B7H6, which encodes an activating ligand for the NK cell receptor NKp30 (Fig. 1C) (30). Another hit from the screen was PVR/CD155, which encodes a ligand for DNAX accessory molecule 1 (DNAM-1), another activating receptor on NK cells (Fig. 1C) (1012). Our screen also recovered IFNGR2, which encodes the interferon- (IFN-) receptor critical to NK cellmediated cytotoxicity (1012). Recovery of these known mediators of NK cell cytotoxicity strongly supports the physiological relevance of the screen and further demonstrates that TALL-104 cells resemble NK cells in tumor killing. Most of the hits, however, were not previously linked to CL-mediated killing.

Multiple genes identified in the screen, including PIGP, PIGS, PIGL, and GPAA1, encode enzymes responsible for the biosynthesis of the GPI moiety of GPI-anchored proteins (GPI-APs) (Fig. 1C) (28). Thus, the GPI anchor biosynthetic pathway is required for tumor cell sensitivity to TALL-104mediated cytotoxicity. Another top-ranking hit was PBRM1/BAF180, which encodes a subunit of the PBAF form of the SWI/SNF chromatin-remodeling complex (Fig. 1C) (31, 32). Other genes isolated in the screen encode factors involved in a range of cellular processes including membrane trafficking and gene expression (fig. S3).

Since a large fraction of the inactivating gene-trap insertions in the primary screen were within the GPI anchor biosynthetic genes (table S1), additional regulators might have been masked. To this end, we next performed a genome-scale modifier screen using GPI-deficient HAP1 cells. We used CRISPR-Cas9 genome editing to delete PIGP, a key gene in GPI anchor biosynthesis (28). Surface expression of the GPI-AP CD59 was abolished (Fig. 2A), confirming the loss of the GPI anchor in PIGP knockout (KO) cells. GPI anchordeficient HAP1 cells were still killed by TALL-104 cells, albeit with a significantly lower efficiency.

(A) Flow cytometry measurements of the surface levels of CD59, a GPI-AP used as a marker for the GPI pathway (CD59 is not involved in CL-mediated killing). PIGP KO HAP1 cells were generated using CRISPR-Cas9 genome editing. A lack of surface CD59 in pooled PIGP KO cells indicates that the GPI pathway was abolished. WT, wild-type. (B) Screen hits from the modifier haploid genetic screen using the PIGP KO HAP1 cells. The screen was performed as described in Fig. 1. The y axis depicts the log10 of P values for the gene hits in the TALL-104 selected population compared to the unselected control using Fishers exact test. We set a P value cutoff of 1 105 to account for multiple hypothesis testing. In addition, we considered a gene as a hit only if it also had strong enrichment for sense-strand intron insertions based on the binomial test (P value cutoff: 1 105). The x axis depicts the chromosomal positions of the genes. The size of the circle is scaled according to the number of unique inactivating gene-trap insertions with the gene. Circles are colored according to the annotated or predicted functions of the gene products. Dashed line indicates the cutoff of significance. Genes that did not reach the cutoff are shown in gray. Full datasets are included in table S2. (C) Lists of genes identified in the primary and modifier screens. Genes linked to cancer or CL killing are highlighted in bold. Cancer association is based on the COSMIC database.

The modifier screen using GPI anchordeficient cells recovered most of the hits from the primary screen including NCR3LG1, PVR, and IFNGR2 (Fig. 2, B and C), validating the critical roles of these genes in TALL-104mediated killing. As expected, none of the genes involved in GPI anchor biosynthesis were recovered in the modifier screen (Fig. 2, B and C; figs. S3 and S4; and table S2). The modifier screen identified many additional genes including TNFRSF10A and TNFRSF10B (Fig. 2, B and C), which encode TRAIL death receptors known to mediate CL-mediated killing (6, 10). These findings are consistent with roles of death receptors in NK cellmediated killing. Other genes identified in the modifier screen were not previously linked to tumor killing. Notably, a subset of the genes identified in the primary screen including PBRM1 were not recovered in the modifier screen (Fig. 2, B and C, and fig. S4), suggesting that these genes might be linked to the GPI biosynthetic pathway in TALL-104mediated killing.

Previous genetic screens dissected MHC-restricted tumor killing mediated by cytotoxic T lymphocytes (3335). Next, we compared the hits from our screens with those from T cellbased screens. The IFN- receptor was also recovered in T cell screens (3335), in agreement with the role of the IFN- pathway in both MHC-restricted and MHC-unrestricted cytotoxicity (8, 10, 12). However, most of the hits from our genetic screens, including the GPI biosynthetic genes, were not isolated in T cellbased screens (3335). Likewise, most genes identified in T cellbased screens were not recovered in our screens. In particular, a large portion of genes identified in T cellbased screens encode regulators of the MHC class I and PD-L1 (programmed cell death 1 ligand 1) pathways (3335). As expected, these genes were not recovered in our MHC-unrestricted screens (Fig. 2C and fig. S4). Notably, although PBRM1 was also isolated in T cellbased screens of tumor killing (35), its function is distinct: PBRM1 inhibits T cellmediated cytotoxicity but promotes TALL-104mediated killing (Fig. 1C) (35). Thus, PBRM1 plays both positive and negative roles in tumor killing. Together, these findings demonstrate differences between the regulatory networks underlying MHC-restricted and MHC-unrestricted cytotoxicity.

According to gene set enrichment analysis (GSEA) (36), the factors identified in our screens belong to multiple KEGG (Kyoto Encyclopedia of Genes and Genomes) pathways and form physical or functional networks (figs. S5 and S6 and table S3). Approximately half of the hits identified in our screens were mutated in human cancers according to the Catalogue of Somatic Mutations in Cancer (COSMIC) database (Fig. 2C). Since these genes do not regulate cell growth (table S4), our data suggest that cancer-linked mutations in these genes may promote tumorigenesis by preventing NK cellmediated tumor killing.

Next, we sought to identify tumor-intrinsic genes encoding negative regulators of TALL-104mediated killing on the basis of depletion of inactivating gene-trap insertions. To accurately calculate gene-trap depletion from a large mutant cell population, we developed gene-trap strand profile (GSP), a scoring metric based on the numbers of unique inactivating (sense) or neutral (antisense) gene-trap insertions within the introns of a candidate gene. A negative GSP score represents the depletion of inactivating gene-trap insertions in a gene, corresponding to a detrimental effect of the gene KO on cell viability (fig. S7, A to C). As a proof of concept, we used the GSP scoring metric to identify the genes essential to the viability or growth of HAP1 cells in the absence of TALL-104 cells. Approximately 1788 essential genes were identified, encompassing all known essential pathways such as translation, transcription, and RNA splicing (fig. S7, D and E, and table S4). This list included virtually all the essential genes identified in previous genetic studies (26, 37), suggesting that GSP scores accurately quantify gene-trap depletions.

Subsequently, we identified genes with significantly negative GSP scores in TALL-104selected HAP1 cells but not in the passage control population. Mutations of these genes sensitize the tumor cells to TALL-104mediated killing, leading to the depletion of inactivating gene-trap insertions. Thus, these genes encode negative regulators of tumor killing by TALL-104 cells. One of the negative factors identified in both the primary and modifier screens was CFLAR (CASP8 and Fas-associated death domain protein (FADD)like apoptosis regulator) (Fig. 3A), a known inhibitor of TRAIL-mediated apoptosis (38, 39). Another negative factor identified in both the primary and modifier screens was MGA, a transcription factor not previously linked to CL-mediated cytotoxicity (Fig. 3A). The ratio of inactivating and neutral gene-trap insertions in CFLAR and MGA genes was approximately 1:1 in the absence of TALL-104 selection, indicating that CFLAR and MGA are not essential to cell growth (Fig. 3A). However, their inactivating gene-trap insertions were strongly depleted after TALL-104 selection (Fig. 3A), reflecting the inhibitory roles of these genes in TALL-104mediated killing. Other negative regulators identified in the screens encompass a range of biological pathways such as TRAIL signaling, transcriptional regulation, and membrane trafficking (Fig. 3, B and C). A group of negative regulators identified in the primary screen were also recovered in the modifier screen (Fig. 3B and fig. S8). Many others, however, were not isolated in the modifier screen (fig. S8 and tables S5 and S6). While a subset of the negative regulators such as CFLAR were also involved in T cellmediated killing (3335), most of the factors were not recovered in T cellbased screens of tumor killing, again highlighting the fundamental differences between MHC-unrestricted and MHC-restricted cytotoxicity.

(A) Visual representations of gene-trap insertions in CFLAR and MGA in the passage control (no TALL-104 selection), the primary screen, and the modifier screen. The gray boxes indicate exons, while the gray lines indicate introns. The gray arrows depict the directions of transcription. Chromosomal locations of inactivating, sense gene-trap insertions are shown in red, while chromosomal locations of neutral, antisense insertions are shown in blue. GSP is a new scoring metric based on the numbers of unique sense (inactivating) or antisense (neutral) gene-trap insertion within the introns of a candidate gene. GSP = [log2(S/A) log10(S A)], where S and A represent the numbers of sense and antisense gene-trap insertions, respectively. (B) Top: Color key of the heatmap. Bottom: Heatmap showing the genes with significantly negative GSP scores in both the primary and modifier screens but not in the passage control. The complete lists of genes are included in fig. S8 and tables S5 and S6. The GSP scores of the genes were quantile-normalized. Genes with significantly negative GSP scores in both screens were clustered using the Euclidean distance metric. Dashed gray lines represent the sample mean, while the solid gray lines represent each hits GSP score relative to the sample mean. The color of each bar represents the GSP score. Genes associated with cancer or CL killing are highlighted in bold. Cancer association is based on the COSMIC database. (C) Summary of the negative regulators identified in the primary and modifier screens based on annotated or predicted gene function. Full datasets are shown in tables S5 and S6.

Next, we sought to determine the molecular mechanism of the GPI anchor biosynthetic pathway in tumor killing. Deletion of the PIGP gene in HAP1 cells abrogated surface expression of GPI-APs (Figs. 2A and 4A and fig. S9A). We mixed the PIGP KO cells with wild-type (WT) HAP1 cells and examined their sensitivity to TALL-104mediated cytotoxicity in a competition assay, which accurately measures the proliferation and death of cell populations (40). We observed that the ratio of WT and PIGP KO cells did not change significantly during passage in the absence of TALL-104 cells (Fig. 4, A and B), confirming that PIGP mutations do not affect cell growth. By contrast, PIGP KO cells were strongly enriched after two rounds of TALL-104 treatment (Fig. 4, A and B). These data demonstrate that mutation of the GPI biosynthetic pathway in HAP1 cells causes resistance to TALL-104mediated killing, validating the result of our genetic screens. Likewise, mutation of PIGP in 786-O cells, a human renal cancer cell line (41), caused resistance of the cells to TALL-104mediated killing (Fig. 4C). Thus, the role of the GPI biosynthetic pathway in tumor killing is not limited to HAP1 cells.

(A) A mixed population of WT and PIGP KO HAP1 cells was either untreated or treated with TALL-104 cells. Negative surface staining of CD55, a GPI-AP not involved in CL-mediated killing, was used as a marker for PIGP KO cells. CD55+ WT cells and CD55 PIGP KO cells were analyzed by flow cytometry. (B) Percentage of WT and PIGP KO HAP1 cells in the passage control or TALL-104treated population. (C) Percentage of WT and PIGP KO 786-O cells in the passage control or TALL-104treated population. CD55+ WT cells and CD55 PIGP KO 786-O cells were quantified by flow cytometry after each round of treatment. Data in (B) and (C) are presented as mean values (n = 3). (D) Normalized surface levels of immune regulators on WT and PIGP KO HAP1 cells. (E) TALL-104 degranulation presented as normalized surface levels of LAMP1 on TALL-104 cells. (F) Normalized surface levels of TRAIL on TALL-104 cells. (G) Normalized surface levels of CD69 on TALL-104 cells. Data in (D) to (G) are presented as means SD (n = 3). P values were calculated using Students t test. n.s., P > 0.05. ***P < 0.001.

TALL-104mediated killing closely resembles NK cellmediated cytotoxicity (Figs. 1 and 2) (42). Next, we directly examined how PIGP KO cells respond to NK cellmediated cytotoxicity. Using a propidium iodide (PI) uptake assay, we observed that PIGP KO cells were substantially more resistant to killing by primary human NK cells (fig. S9B). Thus, mutation of the GPI biosynthetic pathway in target cells also impairs NK cellmediated cytotoxicity.

We then tested how the GPI biosynthetic pathway regulates tumor surface molecules required for MHC-unrestricted cytotoxicity. Both TRAIL receptor and PVR (Poliovirus receptor) regulate TALL-104mediated killing (Fig. 2) (15, 16). However, surface levels of these molecules were not reduced in PIGP KO cells (Fig. 4D), indicating that their functions are independent of the GPI biosynthetic pathway. We then examined UL16-binding proteins (ULBPs), ligands for the activating NK cell receptor NKG2D (19). Since multiple ULBP genes were simultaneously expressed in HAP1 cells (Fig. 4D), ULBPs were not recovered in our screens because of functional redundancy. ULBPs are GPI-APs containing a C-terminal GPI anchor, although a subset of ULBPs also have a transmembrane domain (43, 44). It has been suggested that the GPI anchor and transmembrane domain play redundant roles in ULBP surface localization and activation of NKG2D (43). However, we observed that surface expression of ULBPs was completely lost in PIGP KO cells (Fig. 4D). Thus, despite the presence of transmembrane domains, the GPI anchor is essential to surface expression of ULBPs.

Next, we examined how the GPI biosynthetic pathway in tumor cells influences the function of CLs after cell-cell interaction. A major route of CL-mediated killing involves the secretion of the cytolytic molecules granzyme and perforin (7, 10, 11), which occurs when cytolytic granules fuse with the plasma membrane of a CL (8, 45). To measure cytolytic granule secretion, we used a flow cytometrybased degranulation assay to quantify surface levels of lysosomal-associated membrane protein 1 (LAMP1; also known as CD107a), a cytolytic granule protein that is exposed to the cell surface after cytolytic granules fuse with the plasma membrane (Fig. 4E) (45, 46). LAMP1 is a specific marker for degranulation as the lysosome-like cytolytic granule is not involved in the trafficking of other cytotoxic molecules such as IFN- and death receptor ligands (4750). Using this degranulation assay, we observed that TALL-104 cells exhibited little surface LAMP1 expression in the absence of HAP1 cells (Fig. 4E), consistent with a lack of cytolytic granule secretion in unstimulated CLs. Incubation with HAP1 cells, however, strongly increased the surface levels of LAMP1 on TALL-104 cells (Fig. 4E). Notably, PIGP KO HAP1 cells induced minimal LAMP1 externalization on TALL-104 cells (Fig. 4E), demonstrating that these mutant cells were defective in triggering the secretion of cytolytic molecules.

Besides cytolytic molecules, death ligands such as TRAIL are also used by CLs to kill tumor cells (10, 51). Our screens recovered TRAIL receptors (TNFRSF10A and TNFRSF10B) as hits (Fig. 2, B and C). In the absence of target cells, TALL-104 cells exhibited low surface levels of TRAIL (Fig. 4F). Incubation with WT HAP1 cells strongly elevated surface TRAIL on TALL-104 cells, while PIGP KO HAP1 cells were largely defective in inducing TRAIL surface expression (Fig. 4F). We also examined IFN-, a cytokine released by CLs to promote tumor killing. We observed that similar amounts of IFN- were released from NK cells when they were incubated with WT or PIGP KO HAP1 cells (fig. S10). Thus, IFN- release is not dependent on the GPI biosynthetic pathway of tumor cells.

We then determined whether the GPI biosynthetic pathway regulates CL activation. We examined the surface expression of CD69, an activation marker of NK cells (52, 53). We observed that TALL-104 cells displayed low surface CD69 expression in the absence of HAP1 cells (Fig. 4G). Addition of WT HAP1 cells strongly elevated surface levels of CD69 on TALL-104 cells (Fig. 4G). By contrast, PIGP KO HAP1 cells were largely defective in inducing CD69 expression (Fig. 4G). Thus, PIGP is essential to the activation of TALL-104 cells by tumor cells, consistent with its roles in cytolytic granule secretion and TRAIL expression. Together, these data indicate that the GPI biosynthetic pathway of tumor cells is required for the activation and cytotoxic functions of MHC-unrestricted CLs, providing a molecular explanation for its activating role in MHC-unrestricted cytotoxicity.

We next characterized PBRM1, a subunit of the PBAF chromatin-remodeling complex mutated in a large number of human cancers (Figs. 2 and 5A) (31, 32, 54). To validate the functional role of PBRM1 in TALL-104mediated cytotoxicity, we deleted the PBRM1 gene in HAP1 cells using CRISPR-Cas9 (Fig. 5B). WT and PBRM1 KO HAP1 cells were mixed and subjected to multiple rounds of TALL-104 treatment in a competition assay. We observed that PBRM1 KO cells were significantly enriched after three rounds of TALL-104 treatment (Fig. 5, C and D). By contrast, the ratio of WT and PBRM1 KO cells did not change significantly during passage in the absence of TALL-104 selection (Fig. 5, C and D), confirming that PBRM1 KO did not affect cell growth (32, 55). These results demonstrate that deletion of the PBRM1 gene in target cells caused resistance to TALL-104mediated killing, validating the finding of our genetic screen. Next, we examined the sensitivity of the cells to primary human NK cells. While WT HAP1 cells were efficiently killed by NK cells, PBRM1 KO cells were substantially more resistant to killing (Fig. 5E), confirming the critical role of PBRM1 in NK cellmediated cytotoxicity.

(A) Visual representations of gene-trap insertions in the PBRM1 gene in the screens. The gray boxes indicate exons, and the gray lines indicate introns. The gray arrows depict the direction of transcription. (B) Immunoblot showing PBRM1 expression in WT HAP1 cells and a clonal PBRM1 KO HAP1 cell line. PBRM1 mRNA expression in these cells is shown in fig. S13. (C) WT (GFP+) and PBRM1 KO (GFP) cells in mixed populations were quantified by flow cytometry after three rounds of passage or TALL-104 treatment. (D) Percentage of WT and PBRM1 KO HAP1 cells in the passage control or TALL-104treated population. Data are presented as mean values (n = 3). (E) Percentage of propidium iodidepositive WT and PBRM1 KO HAP1 cells after treatment with primary human NK cells. (F) Mean surface levels of immune regulators on WT and PBRM1 KO HAP1 cells. (G) TALL-104 degranulation presented as normalized surface levels of LAMP1 on TALL-104 cells after incubation with target cells. (H) NK-92 degranulation presented as normalized surface levels of LAMP1 in NK-92 cells. Data in (E) to (H) are presented as means SD (n = 3). P values were calculated using Students t test. n.s., P > 0.05. *P < 0.05, **P < 0.01, ***P < 0.001.

Next, we further examined how PBRM1 regulates the response of tumor cells to killing. Since loss of MHC class I promotes killing by MHC-unrestricted CLs (19), the resistance of PBRM1 KO cells to killing could be due to up-regulation of MHC class I. However, we observed that surface levels of MHC class I molecules were moderately reduced in PBRM1 KO cells (fig. S11), indicating that the resistance of the KO cells to killing was not caused by up-regulation of MHC class I. Surface levels of TRAIL receptor and PVR were not significantly reduced in the KO cells (Fig. 5F). Mutation of PBRM1 markedly decreased surface expression of ULBPs in HAP1 cells (Fig. 5F). Thus, like PIGP, PBRM1 broadly regulates surface expression of ULBPs in target cells. The overall similarity of PIGP and PBRM1 KO phenotypes prompted us to examine whether PBRM1 regulates cytolytic granule secretion. Using the flow cytometrybased degranulation assay, we observed that HAP1-triggered externalization of LAMP1 in TALL-104 cells was markedly reduced when PBRM1 was mutated (Fig. 5G). Likewise, KO of PBRM1 diminished the ability of HAP1 cells to induce LAMP1 externalization in NK cells (Fig. 5H). Thus, PBRM1 is required for triggering cytolytic granule secretion from MHC-unrestricted CLs. We also tested other molecules of CLs involved in target cell killing. We observed that target cellinduced surface expression of TRAIL in TALL-104 cells was reduced when PBRM1 was mutated in HAP1 cells (fig. S12). By contrast, target cellinduced IFN- release was not affected by PBRM1 KO in HAP1 cells (fig. S10). Together, these results demonstrate that PBRM1 regulates ULBP expression in tumor cells and promotes cytolytic granule secretion from MHC-unrestricted CLs.

This work revealed a complex network of tumor-intrinsic factors that positively or negatively regulate the response of tumor cells to NK cells and other clinically significant MHC-unrestricted CLs. Mutations of these factors either cause resistance or enhance sensitivity to killing. The screens identified known genes involved in NK cellmediated killing such as ligands of NK cell receptors. Most of the genes isolated in the screens, however, were not previously linked to tumor killing. Notably, most of our hits including PBRM1 and GPI biosynthetic genes were not recovered in previous attempts to genetically dissect NK cellmediated killing (5658), likely because of the unique genetic screening platform used in this work. The advantages of this platform include homogeneous populations of CLs with consistent tumor-killing activities and a multiround selection procedure that can identify genes with a wide range of KO phenotypes.

A large portion of the genes identified in the screens were mutated in human cancers. Most of these genes do not regulate cell proliferation or response to T cellmediated cytotoxicity, suggesting that their mutations promote cancer progression by impairing NK cellmediated killing of tumor cells. These findings will be valuable for personalized evaluation of cancer genomes and immunotherapeutic strategies. An unexpected discovery of this study is that the genes regulating tumor response to killing are fundamentally distinct between MHC-unrestricted and MHC-restricted CLs. While a subset of our hits including the IFN- receptor and CFLAR are also involved in T cellmediated killing (3335), the vast majority of the genes identified in our screens are unique to tumor response to MHC-unrestricted cytotoxicity. Likewise, most of the genes identified in previous T cellbased screens were not involved in tumor response to killing by MHC-unrestricted CLs. Knowledge of tumor responses to MHC-unrestricted CLs is integral to understanding how tumor cells interact with the immune system and how they evade cancer immunosurveillance to develop into metastatic malignancies.

A major group of genes identified in our screens encode enzymes involved in the biosynthesis of the GPI anchor. We found that mutation of GPI biosynthetic pathway abolishes the surface expression of all ULBPs. This finding indicates that, although certain ULBPs also have transmembrane domains besides the GPI anchor, the GPI anchor is essential for their surface localization. Thus, manipulation of the GPI biosynthetic pathway has the potential to simultaneously target the entire ULBP family of proteins. It is possible that ULBP deficiency fully accounts for the defects of activation and cytotoxic functions of CLs caused by mutations of GPI biosynthesis. However, since cells express nearly 200 GPI-APs (59), it remains possible that additional GPI-APs other than ULBPs are also involved in tumor cells response to killing.

An unexpected finding of this study is that PBRM1 plays opposite roles in MHC-restricted and MHC-unrestricted tumor killing. A tumor suppressor mutated in a number of tumors including clear-cell renal cell carcinoma (31, 32, 35, 54), PBRM1 negatively regulates T cellmediated tumor killing by modulating the expression of cytotoxic signaling molecules (32, 35). While this inhibitory activity correlates well with the clinical benefits of PBRM1 mutations in checkpoint therapies (32, 35), it cannot fully explain the tumor suppressor role of PBRM1. Our findings suggest that PBRM1 mutations promote tumorigenesis by impairing NK cellmediated clearance of tumor cells. PBRM1 regulates surface expression of ULBPs, which are NKG2D ligands. The overall KO phenotype of PBRM1 is remarkably similar to that of PIGP, suggesting that PBRM1 may control the expression, maturation, or localization of a GPI biosynthetic gene(s) rather than individual ULBPs. Further research using human samples and mouse models will be needed to test this possibility.

Both the innate and adaptive immune systems are involved in cancer immunosurveillance, and cancer arises when both systems fail (7, 12). Given the complementary nature of MHC-restricted and MHC-unrestricted CLs in tumor killing, it is conceivable that combinatorial therapies using both types of CLs would achieve the best therapeutic outcomes. NK cells are capable of detecting and eliminating tumor cells refractory to T cellmediated cytotoxicity and are particularly powerful in eradicating metastatic tumor cells and cancer stem cells (1113). To be effective, cancer immunotherapies must overcome resistance to CL-mediated cytotoxicity. The large number of tumor-intrinsic factors identified in this work provides a rich source of potential targets to enhance and maintain the response of tumor cells to endogenous NK cells or engineered NK cells and NK-like cell lines in adoptive transfer therapies.

HAP1 cells were cultured in Iscoves Modified Dulbeccos Medium (IMDM) supplemented with 10% fetal bovine serum (FBS), l-glutamine, and penicillin/streptomycin. 293T cells were cultured in Dulbeccos modified Eagles medium (DMEM) supplemented with 20% FBS, l-glutamine, and penicillin/streptomycin. 786-O cells [American Type Culture Collection (ATCC), no. CRL-1932] were cultured in RPMI 1640 supplemented with 10% FBS, l-glutamine, and penicillin/streptomycin. NK-92 cells (ATCC, no. CRL-2407) were cultured in minimum essential medium Eagle, alpha modification supplemented with 12.5% FBS, 12.5% Donor Equine Serum, 2 mM l-glutamine, 0.2 mM myo-inositol (Acros Organics, no. 122261000), 0.1 mM 2-mercaptoethanol, 0.02 mM folic acid (Acros Organics, no. 216630100), and recombinant human interleukin-1 (IL-2) (100 U/ml) (PeproTech, no. 200-02). TALL-104 cells (ATCC, no. CRL-11386) were cultured in IMDM supplemented with 20% FBS, human albumin (2.5 g/ml) (Sigma-Aldrich, no. A9731), d-mannitol (0.5 g/ml) (Acros Organics, no. 125345000), and recombinant human IL-2 (100 U/ml). Primary NK cells (ZenBio, no. SER-PBCD56 + NK-F) were cultured in RPMI 1640 supplemented with 10% FBS, l-glutamine, penicillin/streptomycin, sodium pyruvate, MEM nonessential amino acids, recombinant human IL-2 (100 U/ml), and recombinant human IL-15 (10 U/ml) (PeproTech, no. 200-15). PBMCs (ZenBio, no. SER-PBMC-200) were cultured in RPMI 1640 supplemented with 10% FBS, l-glutamine, penicillin/streptomycin, sodium pyruvate, and MEM nonessential amino acids. Human ASCs (ZenBio, no. ASC-F) were grown in DMEM/Hams F-12 (1:1) media supplemented with 10% FBS and penicillin/streptomycin.

Mutant HAP1 libraries were produced using a previously described procedure (28). Briefly, gene-trap retroviruses were generated by transfecting six T175 flasks of 293T cells with a cocktail of plasmids including pGT-GFP0, pGT-GFP1, pGT-GFP2, pAdVAntage (Promega, no. E1711), pGAL, and pCMV-VSVG using TurboFectin 8.0 (OriGene, no. TF81001). The retroviruses were collected 40 hours after transfection, and again 50 hours after transfection. The retroviruses were concentrated in a Beckman SW 28 rotor at 25,000 rpm for 1.5 hours. Viral pellets were resuspended in 500 l of phosphate-buffered saline (PBS) overnight at 4C. HAP1 cells (1.5 108) were spin-infected twice at 12-hour intervals with 1.5 viral concentrate (by flask surface area) in the presence of protamine sulfate (8 g/ml). After mixture with virus, cells were plated in 12-well plates at 1.5 106 cells per well and centrifuged at 900g for 1.5 hours at room temperature in a Thermo Fisher Scientific Legend RT+ centrifuge. The multiplicity of infection was kept below 1.0 on the basis of green fluorescent protein (GFP) fluorescence. The GPI-deficient library used in the modifier screen was generated using the above protocol. To produce a mutant library in GPI-APdeficient HAP1 cells, unaltered HAP1 cells were spin-infected with lentiCRISPR virus targeting PIGP (see the Genome editing using CRISPR-Cas9 section). Infected cells were stained for surface CD59 by incubation with anti-CD59 (eBioscience, no. 17-0596) antibodies followed by incubation with allophycocyanin (APC)conjugated anti-mouse secondary antibodies (eBioscience, no. 17-4015) (see the Flow cytometry measurements section). HAP1 cells with low cell size (forward scatter) and lack of surface CD59 protein (APC) were collected by fluorescence-activated cell sorting (MoFlo, Beckman Coulter) to isolate a haploid GPI-AP deficient population. This population was used to produce an additional gene-trap library as described above.

A HAP1 cell library consisting of ~1.5 108 mutagenized cells was thawed and allowed to recover for 3 days. The cells were plated at a density of 5 106 cells per 100-mm plate. One population of 1.5 108 cells was plated for selection, and another population of 1.5 108 cells was plated for passage throughout the duration of the screen. HAP1 cells were incubated with approximately equal numbers of TALL-104 cells in the presence of IL-2 (100 U/ml) and the following inhibitor cocktail: soluble TRAIL-R2-Fc chimera at a final concentration of 20 ng/ml (R&D Systems, no. 631-T2-100), soluble TNFR1p55 at a final concentration of 100 ng/ml (a gift of C. Edwards, University of Colorado School of Medicine), soluble tumor necrosis factor receptor 1 (TNFR1)Fc at a final concentration of 5 ng/ml (R&D Systems, no. 372-RI-050), soluble Fas-Fc chimera at a final concentration of 100 ng/ml (R&D Systems, no. 326-FS-050), and LEAF purified anti-human Fas-L at a final concentration of 50 ng/ml (BioLegend, no. 306409). This inhibitor cocktail was intended to partially inhibit the fast-acting death-receptor killing pathways such that the slower granzyme/perforin cytotoxic pathway could also be reflected in the screen.

The killing and detachment of HAP1 cells were periodically examined until ~50% of the HAP1 cells detached from the plate, which usually took 8 to 12 hours. After treatment, the HAP1 cells were washed once with PBS to remove free TALL-104 cells, and fresh media were added. After 24 hours, the HAP1 cells were dissociated using Accutase (Innovative Cell Technologies, #AT 104), counted, and replated at ~5 106 cells per 100-mm plate for the next round of TALL-104 treatment on the following day. The untreated control cells were split, counted, and replated at a similar total cell number and density as the TALL-104treated HAP1 population. After four rounds of TALL-104 treatment, HAP1 cells were expanded and 5 107 cells were used for genomic DNA extraction and linear polymerase chain reaction (PCR) reactions. The modifier screen using PIGP KO HAP1 cells was completed using a similar protocol as the primary screen, except that HAP1 cells were plated at a density of ~4 106 cells per 100-mm plate and the ratio of TALL-104 to HAP1 cells was approximately 2:1 during treatment.

Genomic DNA was extracted from HAP1 cells using a genomic DNA purification kit (Thermo Fisher Scientific, no. K0721). Linear PCR was performed using 2 g of genomic DNA as template and the following primer: [5-Biotin-GGTCTCCAAATCTCGGTGGAAC-3]. After 125 cycles of amplification, the linear PCR products were purified using biotin-binding Dynabeads (Thermo Fisher Scientific, no. 11047). On-bead ligation of a 5phosphorylated, 3-ddC linker was performed using CircLigase II (Epicentre, no. CL9021K). The product was purified and used as a template for PCR to add Illumina adapter sequences I (5-AATGATACGGCGACCACCGAGATCTGATGGTTCTCTAGCTTGCC-3) and II (5-CAAGCAGAAGACGGCATACGA-3). PCR products from 8 to 30 individual PCR reactions were extracted from a 1% tris-acetate-EDTAagarose gel, pooled over a Qiagen Spin column (Qiagen, no. 28706), and sequenced in a lane of the Illumina HiSeq 2000 using a custom primer recognizing the 5end of the LTR (5-CTAGCTTGCCAAACCTACAGGTGGGGTCTTTCA-3).

FASTQ files from Illumina sequencing were preprocessed to filter duplicate reads using custom scripts. FASTQ files containing unique sequences were aligned to the human genome (hg19) using the Bowtie software v0.12.08 (60). The 50base pair (bp) FASTQ sequences were trimmed from their 3 ends to a length of 35 bp, and were aligned in best mode allowing one mismatch. Reads with more than one genomic alignment were suppressed. Aligned sequences were intersected with gene tables obtained from the University of California, Santa Cruz Genome Browser (hg19) containing either exons or introns using BEDTools software v2.17.0 (61). Unique insertions per gene were counted for exons and for introns. The total numbers of unique sense and antisense insertions within introns were counted for each gene. Fishers exact test was calculated using previously published passage control data (28, 62). We developed custom scripts in Python using NumPy, Pandas, SciPy, and matplotlib modules for downstream data analysis, statistics, and visualization. Heatmaps were generated by normalizing unique insertion counts per gene using the quantile method in R. Normalized values were used as input for heatmap.2 (package gplots) run in R. Insertion plots were produced using the package Gviz in R (www.r-project.org/). GSEA was performed using GSEA from the Broad Institute (36).

Guide sequences were designed according to previously published protocols (63). Double-stranded oligonucleotides containing the guide sequences were individually subcloned into pLentiCRISPR V2-puro vectors as previously described (28, 64). The PIGP gene was mutated using one guide, while the PBRM1 gene was mutated by simultaneous introduction of two independent guide sequences. The following guide sequences were used in this study. The protospacer adjacent motifs are underlined: PIGP: 5-TACAGTACTTTACCTCGTGTGGG-3; PBRM1 exon 1: 5-GAAACCACTTCATAATAGTCTGG-3; PBRM1 exon 4: 5-TTGCAAGCGGCTTTATATTCAGG-3.

Adherent cells were detached from culture plates using Accutase to preserve surface antigens for flow cytometry measurements (6466). Cells were labeled using the indicated mouse monoclonal primary antibodies and APC-conjugated anti-mouse secondary antibodies (eBioscience, no. 17-4015) or Alexa Fluor 488conjugated anti-mouse immunoglobulin G2a secondary antibodies (Invitrogen, #A-21131). Exceptions were phycoerythrin (PE)conjugated anti-CD55 antibodies (eBioscience, no. 12-0559-42), APC-conjugated anti-CD45 antibodies (eBioscience, no. 17-0459-42), APC-conjugated antiHLA-A/B/C antibodies (BioLegend, no. 311409), APC-conjugated anti2-microglobulin antibodies (BioLegend, no. 395711), and eFluor 450conjugated anti-CD69 antibodies (eBioscience, no. 48-0699-41). The following unconjugated primary antibodies were used in this study: anti-CD3 antibodies (eBioscience, no. 14-0038-82), anti-CD56 antibodies (eBioscience, no. 14-0567-82), anti-CD314/NKG2D antibodies (BioLegend, no. 320802), anti-CD337/NKp30 antibodies (BioLegend, no. 325202), anti-PrP antibodies (eBioscience, no. 14-9230), anti-CD59 antibodies (eBioscience, no. 17-0596), anti-CD261/TRAIL-R1 antibodies (eBioscience, no. 14-6644-80), anti-CD262/TRAIL-R2 antibodies (eBioscience, no. 14-9909-82), anti-CD155/PVR antibodies (BioLegend, no. 337602), anti-ULBP1 antibodies (R&D Systems, no. MAB1380), anti-ULBP2/5/6 antibodies (R&D Systems, no. MAB1298), anti-ULBP3 antibodies (R&D Systems, no. MAB1517), anti-LAMP1/CD107a antibodies (eBioscience, no. 14-1079-80), and anti-CD253/TRAIL antibodies (eBioscience, no. 16-9927-82). Flow cytometry data were collected on the CyAn ADP Analyzer (Beckman Coulter). Data from populations of approximately 10,000 cells were analyzed using FlowJo 10.1. All flow cytometry experiments were run in biological triplicate to calculate statistical significance.

Cells were lysed in an SDS sample buffer, and the samples were resolved on 8% bis-tris SDSpolyacrylamide gel electrophoresis. PIGP and PBRM1 were detected using rabbit polyclonal anti-PIGP antibodies (Sino Biological, #204171-T36) and rabbit polyclonal anti-PBRM1/BAF180 antibodies (Bethyl Laboratories, no. A301-591A-T), respectively, and horseradish peroxidaseconjugated anti-rabbit secondary antibodies (Sigma-Aldrich, no. A6154). -Tubulin was probed using mouse monoclonal anti-tubulin antibodies (eBioscience, no. 14-4502-82) and horseradish peroxidaseconjugated anti-mouse secondary antibodies (Sigma-Aldrich, no. A6782).

WT and mutant target cells were mixed at a 1:1 ratio and plated in six-well plates in six replicates at a density of 3.5 105 cells per well. After 24 hours, TALL-104 cells were added to three replicates of target cells at a 1:1 ratio. An additional three replicates were passage controls. After 8 hours, the TALL-104 cells were removed by aspiration and washing with PBS. Target cells attached to the plates were allowed to recover overnight. Subsequently, target cells were collected using Accutase digestion. Approximately half of the collected target cells were plated for an additional round of TALL-104 treatment, while the remaining cells were prepared for flow cytometry measurements. Flow cytometry was used to measure the ratio of WT to mutant target cells in each population.

Target cells were plated at ~105 cells per well in a 24-well plate. After 24 hours, primary human NK cells were stained with APC-conjugated anti-CD45 antibodies and added to target cells at a 1:1 ratio in the presence of IL-2, IL-15, and the inhibitor cocktail. After incubation, both nonadherent and adherent cells were collected using Accutase digestion. The cells were stained with propidium iodide and assayed by flow cytometry.

Target cells were plated at ~105 cells per well in a 24-well plate. After 24 hours, CLs (TALL-104 or NK cells) were added to the target cells at a 1:1 ratio in the presence of IL-2 and the inhibitor cocktail. After incubation for 4 hours, both nonadherent and adherent cells were collected using Accutase digestion. The cells were stained with anti-LAMP1 antibodies (eBioscience, no. 14-1079-80) and PE-conjugated anti-mouse secondary antibodies (BioLegend, no. 406608), as well as APC-conjugated anti-CD45 antibodies (eBioscience, no. 17-0459-42). Only CD45+ CLs were included in the analysis (target cells were CD45). CL degranulation was quantified as the fold change of mean LAMP1 protein surface staining normalized to a CL-only control.

Target cells were plated at ~105 cells per well in a 24-well plate. After 24 hours, CLs (TALL-104 or NK cells) were added to the target cells at a 1:1 ratio in the presence of IL-2. After incubation for 24 hours, both nonadherent and adherent cells were collected using Accutase digestion. The cells were stained with anti-CD253/TRAIL antibodies (eBioscience, no. 16-9927-82), PE-conjugated anti-mouse secondary antibodies, and APC-conjugated anti-CD45 antibodies. TRAIL surface levels on CD45+ CLs were quantified using flow cytometry.

Target cells were plated at ~105 cells per well in a 24-well plate. After 24 hours, CLs were added to the target cells at a 1:1 ratio in the presence of IL-2 and the inhibitor cocktail. After incubation for 48 hours, both nonadherent and adherent cells were collected using Accutase digestion. The cells were stained with eFluor 450conjugated anti-CD69 antibodies (eBioscience, no. 48-0699-41) and APC-conjugated anti-CD45 antibodies. Surface levels of CD69 in CD45+ CLs were quantified using flow cytometry.

Total RNAs were isolated using the RNeasy Mini Kit (Qiagen, no. 74104), followed by treatment with ezDNAse (Thermo Fisher Scientific, no. 18091150). First strand complementary DNA synthesis was performed using a SuperScript IV kit (Thermo Fisher Scientific, no. 18091050). Gene expression was determined by quantitative reverse transcription PCR on a Bio-Rad CFX384 Real-time PCR Detection System using SsoAdvanced Universal SYBR Green Supermix (Bio-Rad, no. 172-5272) with gene-specific primer sets. The cycle threshold values of a candidate gene were normalized to those of GAPDH, a reference gene, and the cycle threshold values were calculated. The results were plotted as fold changes relative to the WT sample.

PCR primers for PBRM1 were as follows: CGGGTGTGATGAACCAAGGA (forward) and

TTGGCTGCTGTATGACAGGG (reverse). PCR primers for GAPDH were as follows: GACAGTCAGCCGCATCTTCT (forward) and GCGCCCAATACGACCAAATC (reverse).

Acknowledgments: We thank C. Dinarello, W. Gao, X. Liu, C. Detweiler, L. Lenz, and B. Weaver for advice or reagents. We thank L. Crisman and I. Datta for comments on the manuscript. Funding: This work was financially supported by grants from NIH (GM126960, AG061829, and DK124431 to J.S.; AI128443 to S.L.; AI135473 to H.Y.; GM088759 to B.L.M. and E.M.D.), a Postdoctoral Fellowship from the Postdoctoral Overseas Training Program at Beijing University of Chinese Medicine (S.W.), and seed grants from the Cancer Center and Linda Crnic Institute at University of Colorado (J.S.). Publication of this article was partially funded by the University of Colorado Boulder Libraries Open Access Fund. Author contributions: B.L.M., E.M.D., and J.S. designed the study. B.L.M., E.M.D., S.W., Y.O., and S.L. performed the experiments. B.L.M., E.M.D., S.W., S.L., H.Y., and J.S. analyzed the results and wrote the manuscript. All the authors contributed to the overall scientific interpretation and edited the manuscript. Competing interests: The authors declare that they have no competing interests. Data and materials availability: All data needed to evaluate the conclusions in the paper are present in the paper and/or the Supplementary Materials. Additional data related to this paper may be requested from the authors. All work was carried out at University of Colorado.

Read this article:
PBRM1 and the glycosylphosphatidylinositol biosynthetic pathway promote tumor killing mediated by MHC-unrestricted cytotoxic lymphocytes - Science...

Read More...

Page 6«..5678..2030..»


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