Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Jeff Hammerbacher is active.

Publication


Featured researches published by Jeff Hammerbacher.


PLOS Medicine | 2017

Contribution of systemic and somatic factors to clinical response and resistance to PD-L1 blockade in urothelial cancer: An exploratory multi-omic analysis

Alexandra Snyder; Tavi Nathanson; Samuel Funt; Arun Ahuja; Jacqueline Buros Novik; Matthew D. Hellmann; Eliza Chang; Bülent Arman Aksoy; Hikmat Al-Ahmadie; Erik Yusko; Marissa Vignali; Sharon Benzeno; Mariel Elena Boyd; Meredith Maisie Moran; Gopa Iyer; Harlan Robins; Elaine R. Mardis; Taha Merghoub; Jeff Hammerbacher; Jonathan E. Rosenberg; Dean F. Bajorin

Background Inhibition of programmed death-ligand 1 (PD-L1) with atezolizumab can induce durable clinical benefit (DCB) in patients with metastatic urothelial cancers, including complete remissions in patients with chemotherapy refractory disease. Although mutation load and PD-L1 immune cell (IC) staining have been associated with response, they lack sufficient sensitivity and specificity for clinical use. Thus, there is a need to evaluate the peripheral blood immune environment and to conduct detailed analyses of mutation load, predicted neoantigens, and immune cellular infiltration in tumors to enhance our understanding of the biologic underpinnings of response and resistance. Methods and findings The goals of this study were to (1) evaluate the association of mutation load and predicted neoantigen load with therapeutic benefit and (2) determine whether intratumoral and peripheral blood T cell receptor (TCR) clonality inform clinical outcomes in urothelial carcinoma treated with atezolizumab. We hypothesized that an elevated mutation load in combination with T cell clonal dominance among intratumoral lymphocytes prior to treatment or among peripheral T cells after treatment would be associated with effective tumor control upon treatment with anti-PD-L1 therapy. We performed whole exome sequencing (WES), RNA sequencing (RNA-seq), and T cell receptor sequencing (TCR-seq) of pretreatment tumor samples as well as TCR-seq of matched, serially collected peripheral blood, collected before and after treatment with atezolizumab. These parameters were assessed for correlation with DCB (defined as progression-free survival [PFS] >6 months), PFS, and overall survival (OS), both alone and in the context of clinical and intratumoral parameters known to be predictive of survival in this disease state. Patients with DCB displayed a higher proportion of tumor-infiltrating T lymphocytes (TIL) (n = 24, Mann-Whitney p = 0.047). Pretreatment peripheral blood TCR clonality below the median was associated with improved PFS (n = 29, log-rank p = 0.048) and OS (n = 29, log-rank p = 0.011). Patients with DCB also demonstrated more substantial expansion of tumor-associated TCR clones in the peripheral blood 3 weeks after starting treatment (n = 22, Mann-Whitney p = 0.022). The combination of high pretreatment peripheral blood TCR clonality with elevated PD-L1 IC staining in tumor tissue was strongly associated with poor clinical outcomes (n = 10, hazard ratio (HR) (mean) = 89.88, HR (median) = 23.41, 95% CI [2.43, 506.94], p(HR > 1) = 0.0014). Marked variations in mutation loads were seen with different somatic variant calling methodologies, which, in turn, impacted associations with clinical outcomes. Missense mutation load, predicted neoantigen load, and expressed neoantigen load did not demonstrate significant association with DCB (n = 25, Mann-Whitney p = 0.22, n = 25, Mann-Whitney p = 0.55, and n = 25, Mann-Whitney p = 0.29, respectively). Instead, we found evidence of time-varying effects of somatic mutation load on PFS in this cohort (n = 25, p = 0.044). A limitation of our study is its small sample size (n = 29), a subset of the patients treated on IMvigor 210 (NCT02108652). Given the number of exploratory analyses performed, we intend for these results to be hypothesis-generating. Conclusions These results demonstrate the complex nature of immune response to checkpoint blockade and the compelling need for greater interrogation and data integration of both host and tumor factors. Incorporating these variables in prospective studies will facilitate identification and treatment of resistant patients.


Cancer immunology research | 2017

Somatic Mutations and Neoepitope Homology in Melanomas Treated with CTLA-4 Blockade

Tavi Nathanson; Arun Ahuja; Alexander Rubinsteyn; Bülent Arman Aksoy; Matthew D. Hellmann; Diana Miao; Eliezer M. Van Allen; Taha Merghoub; Jedd D. Wolchok; Alexandra Snyder; Jeff Hammerbacher

This is a reanalysis of data described in Snyder et al., N Eng J Med 2014;371:2189–99, that also provides an open-source tool for comparing epitopes. No predictor of response to anti–CTLA-4 therapy was more accurate than mutation burden. Immune checkpoint inhibitors are promising treatments for patients with a variety of malignancies. Toward understanding the determinants of response to immune checkpoint inhibitors, it was previously demonstrated that the presence of somatic mutations is associated with benefit from checkpoint inhibition. A hypothesis was posited that neoantigen homology to pathogens may in part explain the link between somatic mutations and response. To further examine this hypothesis, we reanalyzed cancer exome data obtained from our previously published study of 64 melanoma patients treated with CTLA-4 blockade and a new dataset of RNA-Seq data from 24 of these patients. We found that the ability to accurately predict patient benefit did not increase as the analysis narrowed from somatic mutation burden, to inclusion of only those mutations predicted to be MHC class I neoantigens, to only including those neoantigens that were expressed or that had homology to pathogens. The only association between somatic mutation burden and response was found when examining samples obtained prior to treatment. Neoantigen and expressed neoantigen burden were also associated with response, but neither was more predictive than somatic mutation burden. Neither the previously described tetrapeptide signature nor an updated method to evaluate neoepitope homology to pathogens was more predictive than mutation burden. Cancer Immunol Res; 5(1); 84–91. ©2016 AACR.


International Journal of Radiation Oncology Biology Physics | 2016

How Will Big Data Improve Clinical and Basic Research in Radiation Therapy

Barry S. Rosenstein; Jacek Capala; Jason A. Efstathiou; Jeff Hammerbacher; Sarah L. Kerns; Feng Ming Kong; Harry Ostrer; Fred W. Prior; Bhadrasain Vikram; John Wong; Ying Xiao

*Departments of Radiation Oncology, Genetics and Genomic Sciences, Dermatology and Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; yDepartment of Radiation Oncology, New York University School of Medicine, New York, New York; zClinical Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; xDepartment of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; kDepartment of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; {Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York; Department of Radiation Oncology, GRU Cancer Center and Medical College of Georgia, Georgia Regents University, Augusta, Georgia; **Departments of Pathology and Pediatrics, Albert Einstein College of Medicine, Bronx, New York; yyDepartment of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, Arkansas; zzDepartment of Radiation Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland; and xxDepartment of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania


2013 IEEE Symposium on Biological Data Visualization (BioVis) | 2013

invis: Exploring high-dimensional RNA sequences from in vitro selection

Çağatay Demiralp; Eric J. Hayden; Jeff Hammerbacher; Jeffrey Heer

In vitro selection and evolution is a powerful method for discovering RNA molecules based on their binding and catalysis properties. It has important applications to the study of genetic variation and molecular evolution. However, the resulting RNA sequences form a large, high-dimensional space and biologists lack adequate tools to explore and interpret these sequences. We present invis, the first visual analysis tool to facilitate exploration of in vitro selection sequence spaces. invis introduces a novel configuration of coordinated views that enables simultaneous inspection of global projections of sequence data alongside local regions of selected dimensions and sequence clusters. It allows scientists to isolate related sequences for further data analysis, compare sequence populations over varying conditions, filter sequences based on their similarities, and visualize likely pathways of genetic evolution. User feedback indicates that invis enables effective exploration of in vitro RNA selection sequences.


bioRxiv | 2017

PI3Kδ inhibition supports memory T cells with enhanced antitumor fitness

Jacob S. Bowers; Kinga Majchrzak; Michelle H. Nelson; Bülent Arman Aksoy; Megan M. Wyatt; Aubrey S. Smith; Stefanie R. Bailey; Lillian R. Neal; Jeff Hammerbacher; Chrystal M. Paulos

Phosphatidylinositol-3-kinase p110δ (PI3Kδ) inhibition by Idelalisib (CAL-101) in hematological malignancies directly induces apoptosis in cancer cells and disrupts immunological tolerance by depleting regulatory T cells (Tregs). Yet, little is known about the direct impact of PI3Kδ blockade on effector T cells from CAL-101 therapy. Herein, we demonstrate a direct effect of p110δ inactivation via CAL-101 on murine and human CD8+ T cells that promotes a strong undifferentiated memory phenotype (elevated CD62L/CCR7, CD127 and Tcf7). These CAL-101 T cells also persisted longer after transfer and exerted stronger antitumor immunity compared to traditionally expanded CD8+ T cells in two solid tumor models. Thus, this report describes a novel direct enhancement of CD8+ T cell memory by a p110δ inhibitor that leads to markedly improved tumor regression. This finding has significant implications to improve outcomes from next generation cancer immunotherapies. Highlights In vitro blockade of PI3K p110δ with CAL-101 endows antitumor T cells with a stronger memory phenotype than those treated with AKTi The strong memory phenotype of CAL-101 treated cells translates into improved survival of mice bearing aggressive tumors after adoptive transfer of these T cells Human CAR engineered T cells treated with CAL-101 possess an enhanced memory phenotype and robust antitumor efficacy The antitumor efficacy of CAL-101 primed T cells is not mediated by high CD62L or CD127 expression, but is likely driven by their stem memory phenotype eTOC Blurb Bowers et al report a novel function of PI3K blockade using the p110δ subunit inhibitor CAL-101 to induce memory and antitumor potency in CD8+ T cells. Ex vivo treatment of T cells with CAL-101 leads to improved antitumor control and subject survival in both murine transgenic T cell and human CAR T cell models.


bioRxiv | 2017

Contribution of systemic and somatic factors to clinical response and resistance in urothelial cancer: an exploratory multi-omic analysis

Alexandra Snyder; Tavi Nathanson; Samuel Funt; Arun Ahuja; Jacqueline Buros Novik; Matthew D. Hellmann; Eliza Chang; Bülent Arman Aksoy; Hikmat Al-Ahmadie; Erik Yusko; Marissa Vignali; Sharon Benzeno; Mariel Elena Boyd; Meredith Maisie Moran; Gopa Iyer; Harlan Robins; Elaine R. Mardis; Taha Merghoub; Jeff Hammerbacher; Jonathan E. Rosenberg; Dean F. Bajorin

Background: Inhibition of programmed death-ligand one (PD-L1) with atezolizumab can induce durable clinical benefit (DCB) in patients with metastatic urothelial cancers, including complete remissions in patients with chemotherapy refractory disease. Although mutation load and PD-L1 immune cell (IC) staining have been associated with response, they lack sufficient sensitivity and specificity for clinical use. Thus, there is a need to evaluate the peripheral blood immune environment and to conduct detailed analyses of mutation load, predicted neoantigens and immune cellular infiltration in tumors to enhance our understanding of the biologic underpinnings of response and resistance. Methods and Findings: We performed whole exome sequencing (WES), RNA sequencing (RNA-seq), and T cell receptor sequencing (TCR-seq) of pre-treatment tumor samples as well as TCR sequencing of matched, serially collected peripheral blood pre- and post-treatment with atezolizumab. These parameters were assessed for correlation with DCB (defined as progression free survival (PFS) > 6 months) and overall survival (OS), both alone and in the context of clinical and intratumoral parameters known to be predictive of survival in this disease state. Patients with DCB displayed a higher proportion of tumor infiltrating T lymphocytes (TIL) (n=24, Mann-Whitney p=0.047). Pre-treatment peripheral blood TCR clonality below the median was associated with improved PFS (n=29, log-rank p=0.048) and OS (n=29, log-rank p=0.011). Patients with DCB also demonstrated more substantial expansion of tumor-associated TCR clones in the peripheral blood 3 weeks after starting treatment (n=22, Mann-Whitney p=0.022). The combination of high pre-treatment peripheral blood TCR clonality with elevated PD-L1 IC staining in tumor tissue was strongly associated with poor clinical outcomes (n=10, HR=86.22, 95% CI (2.55, 491.65)). Marked variations in mutation loads were seen with different somatic variant calling methodologies, which in turn impacted associations with clinical outcomes. Missense mutation load, predicted neoantigen load and expressed neoantigen load did not demonstrate significant association with DCB (n=25, Mann-Whitney p=0.22, n=25, Mann-Whitney p=0.55, and n=25, Mann-Whitney p=0.29 respectively). Instead, we found evidence of time-varying effects of somatic mutation load on progression-free survival in this cohort (n=25, p=0.044). Conclusions: These results demonstrate the complex nature of immune response to checkpoint blockade and the compelling need for greater interrogation and data integration of both host and tumor factors. Incorporating these variables in prospective studies will facilitate identification and treatment of resistant patients.Background: Inhibition of programmed death-ligand one (PD-L1) with atezolizumab can induce durable clinical benefit (DCB) in patients with metastatic urothelial cancers, including complete remissions in patients with chemotherapy refractory disease. Although mutation load and PD-L1 immune cell (IC) staining have been associated with response, they lack sufficient sensitivity and specificity for clinical use. Thus, there is a need to evaluate the peripheral blood immune environment and to conduct detailed analyses of mutation load, predicted neoantigens and immune cellular infiltration in tumors to enhance our understanding of the biologic underpinnings of response and resistance. Methods and Findings: The goals of this study were to (1) evaluate the association of mutation load and predicted neoantigen load with therapeutic benefit, and (2) determine whether intratumoral and peripheral blood T cell receptor (TCR) clonality inform clinical outcomes in urothelial carcinoma treated with atezolizumab. We hypothesized that an elevated mutation load in combination with T cell clonal dominance among intratumoral lymphocytes prior to treatment or among peripheral T cells after treatment would be associated with effective tumor control upon treatment with anti-PD-L1 therapy. We performed whole exome sequencing (WES), RNA sequencing (RNA-seq), and T cell receptor sequencing (TCR-seq) of pre-treatment tumor samples as well as TCR sequencing of matched, serially collected peripheral blood collected before and after treatment with atezolizumab. These parameters were assessed for correlation with DCB (defined as progression free survival (PFS) > 6 months), PFS, and overall survival (OS), both alone and in the context of clinical and intratumoral parameters known to be predictive of survival in this disease state. Patients with DCB displayed a higher proportion of tumor infiltrating T lymphocytes (TIL) (n=24, Mann-Whitney p=0.047). Pre-treatment peripheral blood TCR clonality below the median was associated with improved PFS (n=29, log-rank p=0.048) and OS (n=29, log-rank p=0.011). Patients with DCB also demonstrated more substantial expansion of tumor-associated TCR clones in the peripheral blood 3 weeks after starting treatment (n=22, Mann-Whitney p=0.022). The combination of high pre-treatment peripheral blood TCR clonality with elevated PD-L1 IC staining in tumor tissue was strongly associated with poor clinical outcomes (n=10, HR (mean)=89.88, HR (median)=23.41, 95% CI (2.43, 506.94), p(HR>1)=0.0014). Marked variations in mutation loads were seen with different somatic variant calling methodologies, which in turn impacted associations with clinical outcomes. Missense mutation load, predicted neoantigen load and expressed neoantigen load did not demonstrate significant association with DCB (n=25, Mann-Whitney p=0.22, n=25, Mann-Whitney p=0.55, and n=25, Mann-Whitney p=0.29 respectively). Instead, we found evidence of time-varying effects of somatic mutation load on progression-free survival in this cohort (n=25, p=0.044). A limitation of our study is its small sample size (n=29), a subset of the patients treated on IMvigor 210 (NCT02108652). Given the number of exploratory analyses performed, we intend for these results to be hypothesis-generating. Conclusions: These results demonstrate the complex nature of immune response to checkpoint blockade and the compelling need for greater interrogation and data integration of both host and tumor factors. Incorporating these variables in prospective studies will facilitate identification and treatment of resistant patients.


F1000Research | 2016

Personalized neoantigen vaccination with synthetic long peptides

John P. Finnigan; Alex Rubinsteyn; Tavi Nathanson; Nicholas Akers; Nina Bhardwaj; Jeff Hammerbacher; Bojan Losic; Eric E. Schadt

§ All non-synonymous somatic variants identified via WES translated into a 40+ character long string corresponding to the amino acid sequence of the mutated genetic region. § Every string is broken into multiple 8-11 character long overlapping substrings, each of which is assessed for pMHC binding affinity, and immunogenicity using NetMHCcons. § Mutated amino acid sequences are ranked based on the sum of putative epitope scores contained in their sequence, and 31-mer vaccine peptides are chosen from the twenty highest scoring sequences via sliding window optimization. § Among all 31-mer sliding windows with equivalent mutant epitope content (candidate vaccine peptides), we select the vaccine peptide with the least number of wildtype epitopes. § The mutation in each vaccine peptide must be at least 5 residues from either the beginning or end of the peptide sequence. References


F1000Research | 2015

Neoantigen homology and predicting response to immune checkpoint blockade in cancer

Arun Ahuja; Tavi Nathanson; Alex Rubinsteyn; Alexandra Snyder; Matt Hellman; Timothy A. Chan; Taha Merghoub; Jedd D. Wolchok; Jeff Hammerbacher

● Pathogen Homology ○ Predicted neoantigens were aligned with T-cell positive peptides from IEDB of the same length, considering positions 3 through n-1 (n = length) ○ Peptide alignment was scored with the PMBEC matrix [3] and a gap penalty of min(PMBEC). For example, the following entry had a score of 1.4: Immune checkpoint inhibitors are promising cancer treatments for a variety of malignancies, but accurate prediction of clinical response remains an active area of research.


Archive | 2009

Beautiful Data: The Stories Behind Elegant Data Solutions

Jeff Hammerbacher; Egon Willighagen; Peter Norvig; Yair Ghitza; Toby Segaran; Jean-Claude Bradley; Matthew D. Wood; Cameron Neylon; Ben Blackburne; Jeff Jonas; Nathan Yau; Lisa Sokol; Jason Dykes; David Poole; Andrew Gelman; Antony Williams; Brian F. Cooper; Jud Valeski; Jonathan P. Kastellec; Matthew Holm; Andrew S. I. D. Lang; Jeffrey Heer; Jayant Madhavan; Deborah F. Swayne; Valdean Klump; Pierre Lindenbaum; Brendan O'Connor; Lukas Biewald; Rajershi Guha; Alon Y. Halevy


international conference on management of data | 2015

Rethinking Data-Intensive Science Using Scalable Analytics Systems

Frank Austin Nothaft; Matt Massie; Timothy Danford; Zhao Zhang; Uri Laserson; Carl Yeksigian; Jey Kottalam; Arun Ahuja; Jeff Hammerbacher; Michael D. Linderman; Michael J. Franklin; Anthony D. Joseph; David A. Patterson

Collaboration


Dive into the Jeff Hammerbacher's collaboration.

Top Co-Authors

Avatar

Arun Ahuja

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Tavi Nathanson

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Alex Rubinsteyn

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Alexandra Snyder

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Bülent Arman Aksoy

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Taha Merghoub

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jacqueline Buros Novik

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

John P. Finnigan

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Matthew D. Hellmann

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge