80 results on '"Jeffrey Chou"'
Search Results
2. 778 TILVANCE-301, a phase 3 study of lifileucel tumor-infiltrating lymphocyte (TIL) cell therapy combined with pembrolizumab (pembro) vs pembro alone in treatment-naïve unresectable or metastatic melanoma
- Author
-
Gino K In, James Larkin, Yazan Samhouri, Marcus Butler, Victoria Atkinson, Jeffrey Chou, Juan Martin-Liberal, Patrick Terheyden, Sajeve Thomas, Wen Shi, Xiao Wu, Andrew S Poklepovic, Young Hong, Giri Sulur, Friedrich Graf Finckenstein, John B Haanen, Andrew JS Furness, Philip Lammers, Ryan H Nguyen, and Idit Peretz
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
3. First-in-human study of an OX40 (ivuxolimab) and 4-1BB (utomilumab) agonistic antibody combination in patients with advanced solid tumors
- Author
-
Toshihiko Doi, John A Thompson, Omid Hamid, Adi Diab, Siwen Hu-Lieskovan, Jeffrey Chou, Jeffrey S Wasser, Patrick A Ott, Jean-Philippe Spano, Willeke Ros, Alison Forgie, Wenjing Yang, Ferry A L M Eskens, Alberto A Chiappori, Eric Angevin, Naiyer A Rizvi, Cen Guo, and Anthony B El-Khoueiry
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background Ivuxolimab (PF-04518600) and utomilumab (PF-05082566) are humanized agonistic IgG2 monoclonal antibodies against OX40 and 4-1BB, respectively. This first-in-human, multicenter, open-label, phase I, dose-escalation/dose-expansion study explored safety, tolerability, pharmacokinetics, pharmacodynamics, and antitumor activity of ivuxolimab+utomilumab in patients with advanced solid tumors.Methods Dose-escalation: patients with advanced bladder, gastric, or cervical cancer, melanoma, head and neck squamous cell carcinoma, or non-small cell lung cancer (NSCLC) who were unresponsive to available therapies, had no standard therapy available or declined standard therapy were enrolled into five dose cohorts: ivuxolimab (0.1–3 mg/kg every 2 weeks (Q2W)) intravenously plus utomilumab (20 or 100 mg every 4 weeks (Q4W)) intravenously. Dose-expansion: patients with melanoma (n=10) and NSCLC (n=20) who progressed on prior anti-programmed death receptor 1/programmed death ligand-1 and/or anti-cytotoxic T-lymphocyte-associated antigen 4 (melanoma) received ivuxolimab 30 mg Q2W intravenously plus utomilumab 20 mg Q4W intravenously. Adverse events (AEs) were graded per National Cancer Institute Common Terminology Criteria for Adverse Events V.4.03 and efficacy was assessed using Response Evaluation Criteria in Solid Tumors (RECIST) V.1.1 and immune-related RECIST (irRECIST). Paired tumor biopsies and whole blood were collected to assess pharmacodynamic effects and immunophenotyping. Whole blood samples were collected longitudinally for immunophenotyping.Results Dose-escalation: 57 patients were enrolled; 2 (3.5%) patients with melanoma (0.3 mg/kg+20 mg and 0.3 mg/kg+100 mg) achieved partial response (PR), 18 (31.6%) patients achieved stable disease (SD); the disease control rate (DCR) was 35.1% across all dose levels. Dose-expansion: 30 patients were enrolled; 1 patient with NSCLC achieved PR lasting >77 weeks. Seven of 10 patients with melanoma (70%) and 7 of 20 patients with NSCLC (35%) achieved SD: median (range) duration of SD was 18.9 (13.9–49.0) weeks for the melanoma cohort versus 24.1 (14.3–77.9+) weeks for the NSCLC cohort; DCR (NSCLC) was 40%. Grade 3–4 treatment-emergent AEs were reported in 28 (49.1%) patients versus 11 (36.7%) patients in dose-escalation and dose-expansion, respectively. There were no grade 5 AEs deemed attributable to treatment. Ivuxolimab area under the concentration–time curve increased in a dose-dependent manner at 0.3–3 mg/kg doses.Conclusions Ivuxolimab+utomilumab was found to be well tolerated and demonstrated preliminary antitumor activity in selected groups of patients.Trial registration number NCT02315066.
- Published
- 2022
- Full Text
- View/download PDF
4. Immunogenicity of immunomodulatory, antibody-based, oncology therapeutics
- Author
-
Jasmine Davda, Paul Declerck, Siwen Hu-Lieskovan, Timothy P. Hickling, Ira A. Jacobs, Jeffrey Chou, Shahram Salek-Ardakani, and Eugenia Kraynov
- Subjects
ADA ,Antibody ,Immunogenicity ,Immunomodulatory ,Neutralizing antibodies ,PD-1/PD-L1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract The increasing use of multiple immunomodulatory (IMD) agents for cancer therapies (e.g. antibodies targeting immune checkpoints, bispecific antibodies, and chimeric antigen receptor [CAR]-T cells), is raising questions on their potential immunogenicity and effects on treatment. In this review, we outline the mechanisms of action (MOA) of approved, antibody-based IMD agents, potentially related to their immunogenicity, and discuss the reported incidence of anti-drug antibodies (ADA) as well as their clinical relevance in patients with cancer. In addition, we discuss the impact of the administration route and potential strategies to reduce the incidence of ADA and manage treated patients. Analysis of published reports indicated that the risk of immunogenicity did not appear to correlate with the MOA of anti-programmed death 1 (PD-1)/PD-ligand 1 monoclonal antibodies nor to substantially affect treatment with most of these agents in the majority of patients evaluated to date. Treatment with B-cell depleting agents appears associated with a low risk of immunogenicity. No significant difference in ADA incidence was found between the intravenous and subcutaneous administration routes for a panel of non-oncology IMD antibodies. Additionally, while the data suggest a higher likelihood of immunogenicity for antibodies with T-cell or antigen-presenting cell (APC) targets versus B-cell targets, it is possible to have targets expressed on APCs or T cells and still have a low incidence of immunogenicity.
- Published
- 2019
- Full Text
- View/download PDF
5. Phenotypic and transcriptional fidelity of patient-derived colon cancer xenografts in immune-deficient mice.
- Author
-
Jeffrey Chou, Matthew P Fitzgibbon, Christie-Lynn L Mortales, Andrea M H Towlerton, Melissa P Upton, Raymond S Yeung, Martin W McIntosh, and Edus H Warren
- Subjects
Medicine ,Science - Abstract
Xenografts of human colorectal cancer (CRC) in immune-deficient mice have great potential for accelerating the study of tumor biology and therapy. We evaluated xenografts established in NOD/scid/IL2Rγ-null mice from the primary or metastatic tumors of 27 patients with CRC to estimate their capacity for expanding tumor cells for in vitro studies and to assess how faithfully they recapitulated the transcriptional profile of their parental tumors. RNA-seq analysis of parental human CRC tumors and their derivative xenografts demonstrated that reproducible transcriptional changes characterize the human tumor to murine xenograft transition. In most but not all cases, the human stroma, vasculature, and hematopoietic elements were systematically replaced by murine analogues while the carcinoma component persisted. Once established as xenografts, human CRC cells that could be propagated by serial transplantation remained transcriptionally stable. Three histologically atypical xenografts, established from patients with peritoneal metastases, contained abundant human stromal elements and blood vessels in addition to human tumor cells. The transcriptomes of these mixed tumor/stromal xenografts did not closely resemble those of their parental tumors, and attempts to propagate such xenografts by serial transplantation were unsuccessful. Stable expression of numerous genes previously identified as high priority targets for immunotherapy was observed in most xenograft lineages. Aberrant expression in CRC cells of human genes that are normally only expressed in hematopoietic cells was also observed. Our results suggest that human CRC cells expanded in murine xenografts have great utility for studies of tumor immunobiology and targeted therapies such as immunotherapy but also identify potential limitations.
- Published
- 2013
- Full Text
- View/download PDF
6. NYESO-1/LAGE-1s and PRAME are targets for antigen specific T cells in chondrosarcoma following treatment with 5-Aza-2-deoxycitabine.
- Author
-
Seth M Pollack, Yonqing Li, Megan J Blaisdell, Erik A Farrar, Jeffrey Chou, Benjamin L Hoch, Elizabeth T Loggers, Eve Rodler, Janet F Eary, Ernest U Conrad, Robin L Jones, and Cassian Yee
- Subjects
Medicine ,Science - Abstract
Chondrosarcoma has no proven systemic option in the metastatic setting. The development of a non-cross-resistant strategy, such as cellular immunotherapy using antigen-specific T cells would be highly desirable. NY-ESO-1 and PRAME are members of the Cancer Testis Antigen (CTA) family that have been identified as promising targets for T cell therapy. LAGE-1 is a cancer testis antigen 90% homologous to NY-ESO-1, sharing the 157-165 A*0201 NY-ESO-1 epitope with its transcript variant, LAGE-1s. A number of CTA's have been induced using 5-Aza-2-Deoxycitabine (5-Aza-dC) in other cancers. We sought to evaluate the feasibility of targeting chondrosarcoma tumors using NY-ESO-1/LAGE-1s and PRAME specific T cells using 5-Aza-dC to induce antigen expression.We used 11 flash frozen tumors from the University of Washington tumor bank to test for the expression of NY-ESO-1, PRAME, LAGE-1s and LAGE-1L in chondrosarcoma tumors. Using four chondrosarcoma cell lines we tested the expression of these CTA's with and without 5-Aza-dC treatments. Finally, using NY-ESO-1/LAGE-1s and PRAME specific effectors that we generated from sarcoma patients, we evaluated the ability of these T cells to lyse A*0201 expressing chondrosarcoma cell lines in vitro both with and without 5-Aza-dC treatment.A minority (36%) of chondrosarcoma tumors expressed either NY-ESO-1 or LAGE-1s at >10% of our reference value and none expressed PRAME at that level. However, in all four of the chondrosarcoma cell lines tested, NY-ESO-1 and PRAME expression could be induced following treatment with 5-Aza-dC including in cell lines where expression was absent or barely detectable. Furthermore, NY-ESO-1/LAGE-1s and PRAME specific CD8+ effector T cells were able to specifically recognize and lyse A*0201 expressing chondrosarcoma cell lines following 5-Aza-dC treatment.These data suggest that adoptive immunotherapy in combination with 5-Aza-dC may be a potential strategy to treat unresectable or metastatic chondrosarcoma patients where no proven systemic therapies exist.
- Published
- 2012
- Full Text
- View/download PDF
7. Figure S10 from Pharmacologic Properties and Preclinical Activity of Sasanlimab, A High-affinity Engineered Anti-Human PD-1 Antibody
- Author
-
Shahram Salek-Ardakani, John C. Lin, Arvind Rajpal, Eugenia Kraynov, Jeffrey Chou, Jerry D. Clark, Allison Xu, Christopher P. Dillon, Natalija Budimir, Brent Kern, Bart Jessen, Cris Kamperschroer, Sherman M. Chin, Christopher R. Kimberlin, Joyce Chou, HoangKim Nguyen, Yasmina Abdiche, Sawsan Youssef, and Amir A. Al-Khami
- Abstract
Supplementary Figure 10 shows the selectivity of sasanlimab binding to human and cynomolgus monkey PD-1 using HEK-293T transiently transfected cells and flow cytometry
- Published
- 2023
8. Data from Pharmacologic Properties and Preclinical Activity of Sasanlimab, A High-affinity Engineered Anti-Human PD-1 Antibody
- Author
-
Shahram Salek-Ardakani, John C. Lin, Arvind Rajpal, Eugenia Kraynov, Jeffrey Chou, Jerry D. Clark, Allison Xu, Christopher P. Dillon, Natalija Budimir, Brent Kern, Bart Jessen, Cris Kamperschroer, Sherman M. Chin, Christopher R. Kimberlin, Joyce Chou, HoangKim Nguyen, Yasmina Abdiche, Sawsan Youssef, and Amir A. Al-Khami
- Abstract
Development of antagonistic mAbs that specifically target the immune checkpoint receptor, programmed cell death protein-1 (PD-1), is of great interest for cancer immunotherapy. Here, we report the biophysical characteristics and nonclinical antagonistic activities of sasanlimab (PF-06801591), a humanized anti-PD-1 antibody of IgG4 isotype. We show that sasanlimab binds selectively and with similar high potency to human and cynomolgus monkey PD-1 receptor and blocks its interaction with PD-L1 and PD-L2, with no detectable Fc-dependent effector function. The binding of sasanlimab to human and cynomolgus PD-1 is associated with the formation of a stable complex, which is likely to be the main driver of this high-affinity interaction. In vitro, sasanlimab significantly augmented T-cell proliferation and cytokine production in mixed lymphocyte reaction and superantigen stimulation assays. In vivo, sasanlimab accelerated the incidence of GvHD by enhancing T-cell proliferation and cytokine secretion in a xenogeneic model of acute GvHD and halted the growth of MC-38 colon adenocarcinoma tumors in human PD-1 knock-in mice. Pharmacokinetic and toxicokinetic findings from cynomolgus monkey showed that sasanlimab was active and well-tolerated. Taken together, the data presented here support the clinical development of sasanlimab for the treatment of patients with advanced cancers as a single agent or in combination with other immunotherapies.
- Published
- 2023
9. CCR Translation on this Article from C19orf48 Encodes a Minor Histocompatibility Antigen Recognized by CD8+ Cytotoxic T Cells from Renal Cell Carcinoma Patients
- Author
-
Edus H. Warren, Stanley R. Riddell, Benoît Van den Eynde, Peter Cresswell, Brenda M. Sandmaier, John A. Thompson, Lilien N. Voong, Jeffrey Chou, Maureen X. Miranda, Jeffrey K. Mito, Sharon M. Lu, Nathalie Vigneron, Nobuharu Fujii, and Scott S. Tykodi
- Abstract
CCR Translation on this Article from C19orf48 Encodes a Minor Histocompatibility Antigen Recognized by CD8+ Cytotoxic T Cells from Renal Cell Carcinoma Patients
- Published
- 2023
10. Data from C19orf48 Encodes a Minor Histocompatibility Antigen Recognized by CD8+ Cytotoxic T Cells from Renal Cell Carcinoma Patients
- Author
-
Edus H. Warren, Stanley R. Riddell, Benoît Van den Eynde, Peter Cresswell, Brenda M. Sandmaier, John A. Thompson, Lilien N. Voong, Jeffrey Chou, Maureen X. Miranda, Jeffrey K. Mito, Sharon M. Lu, Nathalie Vigneron, Nobuharu Fujii, and Scott S. Tykodi
- Abstract
Purpose: Tumor regression has been observed in some patients with metastatic renal cell carcinoma (RCC) after nonmyeloablative allogeneic hematopoietic cell transplantation (HCT). Cellular and molecular characterization of antigens recognized by tumor-reactive T cells isolated from responding patients could potentially provide insight into the mechanisms of tumor regression.Experimental Design: CD8+ CTL clones that recognized a novel RCC-associated minor histocompatibility (H) antigen presented by HLA-A*0201 were isolated from two patients with metastatic RCC who experienced tumor regression or stable disease following nonmyeloablative allogeneic HCT. These clones were used to screen a cDNA library and isolate the unique cDNA encoding the antigen.Results: An alternative open reading frame in the C19orf48 gene located on chromosome 19q13 encodes the HLA-A*0201–restricted minor H antigen recognized by the RCC-reactive T cells. The differential T-cell recognition of donor- and recipient-derived target cells is attributable to a nonsynonymous single-nucleotide polymorphism within the nucleotide interval that encodes the antigenic peptide. Assays for gene expression and CTL recognition showed that the C19orf48-encoded peptide is widely expressed in renal tumors and solid tumors of other histologies. The antigenic peptide can be processed for CTL recognition via both TAP-dependent and TAP-independent pathways.Conclusions: Donor T-cell responses against the HLA-A*0201–restricted minor H antigen encoded by C19orf48 may contribute to RCC regression after MHC-matched allogeneic HCT.
- Published
- 2023
11. Data from A Phase I, Open-Label, Dose-Escalation Study of the OX40 Agonist Ivuxolimab in Patients with Locally Advanced or Metastatic Cancers
- Author
-
Anthony B. El-Khoueiry, Jeffrey Chou, Carrie Turich Taylor, Shahram Salek-Ardakani, Ken Liao, Tenshang Joh, Xiao Wang, Catherine Fleener, Bishu Ganguly, Samuel J. Klempner, Siwen Hu-Lieskovan, Toshihiko Doi, Ferry A.L.M. Eskens, Willeke Ros, John A. Thompson, Omid Hamid, and Adi Diab
- Abstract
Purpose:Stimulation of effector T cells is an appealing immunotherapeutic approach in oncology. OX40 (CD134) is a costimulatory receptor expressed on activated CD4+ and CD8+ T cells. Induction of OX40 following antigen recognition results in enhanced T-cell activation, proliferation, and survival, and OX40 targeting shows therapeutic efficacy in preclinical studies. We report the monotherapy dose-escalation portion of a multicenter, phase I trial (NCT02315066) of ivuxolimab (PF-04518600), a fully human immunoglobulin G2 agonistic monoclonal antibody specific for human OX40.Patients and Methods:Adult patients (N = 52) with selected locally advanced or metastatic cancers received ivuxolimab 0.01 to 10 mg/kg. Primary endpoints were safety and tolerability. Secondary/exploratory endpoints included preliminary assessment of antitumor activity and biomarker analyses.Results:The most common all-causality adverse events were fatigue (46.2%), nausea (28.8%), and decreased appetite (25.0%). Of 31 treatment-related adverse events, 30 (96.8%) were grade ≤2. No dose-limiting toxicities occurred. Ivuxolimab exposure increased in a dose-proportionate manner from 0.3 to 10 mg/kg. Full peripheral blood target engagement occurred at ≥0.3 mg/kg. Three (5.8%) patients achieved a partial response, and disease control was achieved in 56% of patients. Increased CD4+ central memory T-cell proliferation and activation, and clonal expansion of CD4+ and CD8+ T cells in peripheral blood were observed at 0.1 to 3.0 mg/kg. Increased immune cell infiltrate and OX40 expression were evident in on-treatment tumor biopsies.Conclusions:Ivuxolimab was generally well tolerated with on-target immune activation at clinically relevant doses, showed preliminary antitumor activity, and may serve as a partner for combination studies.
- Published
- 2023
12. Supplementary Data from A Phase I, Open-Label, Dose-Escalation Study of the OX40 Agonist Ivuxolimab in Patients with Locally Advanced or Metastatic Cancers
- Author
-
Anthony B. El-Khoueiry, Jeffrey Chou, Carrie Turich Taylor, Shahram Salek-Ardakani, Ken Liao, Tenshang Joh, Xiao Wang, Catherine Fleener, Bishu Ganguly, Samuel J. Klempner, Siwen Hu-Lieskovan, Toshihiko Doi, Ferry A.L.M. Eskens, Willeke Ros, John A. Thompson, Omid Hamid, and Adi Diab
- Abstract
Supplementary Data from A Phase I, Open-Label, Dose-Escalation Study of the OX40 Agonist Ivuxolimab in Patients with Locally Advanced or Metastatic Cancers
- Published
- 2023
13. Supplementary Figures S1-S2 from C19orf48 Encodes a Minor Histocompatibility Antigen Recognized by CD8+ Cytotoxic T Cells from Renal Cell Carcinoma Patients
- Author
-
Edus H. Warren, Stanley R. Riddell, Benoît Van den Eynde, Peter Cresswell, Brenda M. Sandmaier, John A. Thompson, Lilien N. Voong, Jeffrey Chou, Maureen X. Miranda, Jeffrey K. Mito, Sharon M. Lu, Nathalie Vigneron, Nobuharu Fujii, and Scott S. Tykodi
- Abstract
Supplementary Figures S1-S2 from C19orf48 Encodes a Minor Histocompatibility Antigen Recognized by CD8+ Cytotoxic T Cells from Renal Cell Carcinoma Patients
- Published
- 2023
14. Association of Tumor Mutational Burden and Immune Gene Expression with Response to PD-1 Blockade by Sasanlimab Across Tumor Types and Routes of Administration
- Author
-
Alison Forgie, Melissa Lynne Johnson, Jeffrey Chou, Ira Jacobs, Xiao Wang, Siwen Hu-Lieskovan, Juneko E. Grilley-Olson, Fadi Braiteh, and Vinicius Bonato
- Subjects
Cancer Research ,medicine.drug_class ,Programmed Cell Death 1 Receptor ,Gene Expression ,Monoclonal antibody ,B7-H1 Antigen ,Interferon-gamma ,Neoplasms ,Biomarkers, Tumor ,medicine ,Humans ,Pharmacology (medical) ,Original Research Article ,Receptor ,Immune Checkpoint Inhibitors ,biology ,business.industry ,Antibodies, Monoclonal ,Cell cycle ,Acquired immune system ,Oncology ,biology.protein ,Cancer research ,Biomarker (medicine) ,Immunohistochemistry ,Antibody ,business ,CD8 - Abstract
Background Sasanlimab is a monoclonal antibody that binds to the programmed cell death receptor 1 (PD-1). Anti-PD-1 monoclonal antibodies have improved patient clinical outcomes; however, not all treated patients derive clinical benefit. Further insights on potential biomarkers beyond PD-L1 expression levels would help to identify the patients most likely to respond to treatment. Objective This study evaluated tumor biopsies from patients treated with intravenous or subcutaneous sasanlimab to identify biomarkers of response and characterize pharmacodynamic activity. Methods Anti-PD-1/PD-ligand 1 (PD-L1)-naive patients with advanced solid tumors received sasanlimab intravenously at 1, 3, or 10 mg/kg every 3 weeks (n = 23) or subcutaneously at 300 mg every 4 weeks (n = 15). Best tumor percentage change from baseline was determined by RECIST. Whole-exome DNA and RNA sequencing were performed in tumor samples collected from treated patients at protocol-defined timepoints. PD-L1 and CD8 protein expression were evaluated in tumor biopsies by immunohistochemistry. Associations with response were assessed by linear regression analysis. Results Baseline tumor mutational burden (TMB), as well as PD-L1 and CD8 expression, were significantly associated with response to sasanlimab across the multiple dose levels, routes of administration, and range of tumor types evaluated. TMB is an independent biomarker from the various tumor inflammatory genes and signatures evaluated. Gene set enrichment analysis showed that higher baseline expression levels of genes related to the interferon-γ and PD-1 signaling pathways and the cell cycle were significantly associated with response to sasanlimab across tumor types. No differences were observed between routes of administration with regard to response to sasanlimab for the biomarkers of interest (TMB, PD-L1, CD8, and interferon-γ signature). Evaluation of pharmacodynamic changes showed increased tumor expression of genes enriched in adaptive immune response pathways. Conclusions Our findings indicate an active, immunomodulatory mechanism for the anti-PD-1 antibody sasanlimab across different tumor types and routes of administration. Trial Registration ClinicalTrials.gov identifier NCT02573259; registered October 2015. Supplementary Information The online version contains supplementary material available at 10.1007/s11523-021-00833-2.
- Published
- 2021
15. 727 Interim phase I clinical data of FT538, an off-the-shelf, multiplexed-engineered, iPSC-derived NK cell therapy, combined with monoclonal antibodies in patients with advanced solid tumors
- Author
-
Martin Gutierrez, Melissa Johnson, David Sommerhalder, Wells Messersmith, Haeseong Park, Muhammad Furqan, Jason Chesney, Ryan Bjordahl, Lingmin Zeng, Peter Szabo, Yu-Waye Chu, Brandon Beagle, Bob Valamehr, Jeffrey Chou, and David Hong
- Published
- 2022
16. 755 Results of a phase I trial of FT500, a first-in-class, off-the-shelf, iPSC–derived NK cell therapy combined with PD-1/PD-L1 checkpoint blockade therapy and IL-2 in patients with advanced solid tumors
- Author
-
Sandip Patel, Manish Patel, Martin Gutierrez, Marlisa Anderson, Peter Szabo, Bob Valamehr, Yu-Waye Chu, Brandon Beagle, Jeffrey Chou, and David Hong
- Published
- 2022
17. 726 Phase I results of FT516, an off-the-shelf, iPSC-derived NK cell therapy expressing a high-affinity, non-cleavable CD16 (hnCD16) combined with avelumab in patients with advanced solid tumors
- Author
-
Martin Gutierrez, Manish Patel, Feng Liu, Peter Szabo, Bob Valamehr, Yu-Waye Chu, Brandon Beagle, Jeffrey Chou, and David Hong
- Published
- 2022
18. 204 Combining FT536, a pan-tumor targeting CAR NK cell therapy, with CD16 engagers provides a coordinated targeting strategy to overcome tumor heterogeneity
- Author
-
John Goulding, Bryan Hancock, Robert Blum, Wen-I Yeh, Chia-Wei Chang, Mochtar Pribadi, Yijia Pan, Hui-Yi Chu, Shohreh Sikaroodi, Thomas Dailey, Miguel Meza, Lucas Ferrari de Andrade, Peter Szabo, Sarah Cooley, Jeffrey Chou, John Powderly, Yu-Waye Chu, Tom Lee, Ryan Bjordahl, Kai Wucherpfennig, and Bob Valamehr
- Published
- 2022
19. First-in-human study of an OX40 (ivuxolimab) and 4-1BB (utomilumab) agonistic antibody combination in patients with advanced solid tumors
- Author
-
Omid Hamid, Alberto A Chiappori, John A Thompson, Toshihiko Doi, Siwen Hu-Lieskovan, Ferry A L M Eskens, Willeke Ros, Adi Diab, Jean-Philippe Spano, Naiyer A Rizvi, Jeffrey S Wasser, Eric Angevin, Patrick A Ott, Alison Forgie, Wenjing Yang, Cen Guo, Jeffrey Chou, Anthony B El-Khoueiry, and Medical Oncology
- Subjects
Pharmacology ,Cancer Research ,Lung Neoplasms ,Immunology ,Antibodies, Monoclonal ,Antibodies, Monoclonal, Humanized ,Oncology ,SDG 3 - Good Health and Well-being ,Head and Neck Neoplasms ,Carcinoma, Non-Small-Cell Lung ,Immunoglobulin G ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Molecular Medicine ,Immunology and Allergy ,Melanoma - Abstract
BackgroundIvuxolimab (PF-04518600) and utomilumab (PF-05082566) are humanized agonistic IgG2 monoclonal antibodies against OX40 and 4-1BB, respectively. This first-in-human, multicenter, open-label, phase I, dose-escalation/dose-expansion study explored safety, tolerability, pharmacokinetics, pharmacodynamics, and antitumor activity of ivuxolimab+utomilumab in patients with advanced solid tumors.MethodsDose-escalation: patients with advanced bladder, gastric, or cervical cancer, melanoma, head and neck squamous cell carcinoma, or non-small cell lung cancer (NSCLC) who were unresponsive to available therapies, had no standard therapy available or declined standard therapy were enrolled into five dose cohorts: ivuxolimab (0.1–3 mg/kg every 2 weeks (Q2W)) intravenously plus utomilumab (20 or 100 mg every 4 weeks (Q4W)) intravenously. Dose-expansion: patients with melanoma (n=10) and NSCLC (n=20) who progressed on prior anti-programmed death receptor 1/programmed death ligand-1 and/or anti-cytotoxic T-lymphocyte-associated antigen 4 (melanoma) received ivuxolimab 30 mg Q2W intravenously plus utomilumab 20 mg Q4W intravenously. Adverse events (AEs) were graded per National Cancer Institute Common Terminology Criteria for Adverse Events V.4.03 and efficacy was assessed using Response Evaluation Criteria in Solid Tumors (RECIST) V.1.1 and immune-related RECIST (irRECIST). Paired tumor biopsies and whole blood were collected to assess pharmacodynamic effects and immunophenotyping. Whole blood samples were collected longitudinally for immunophenotyping.ResultsDose-escalation: 57 patients were enrolled; 2 (3.5%) patients with melanoma (0.3 mg/kg+20 mg and 0.3 mg/kg+100 mg) achieved partial response (PR), 18 (31.6%) patients achieved stable disease (SD); the disease control rate (DCR) was 35.1% across all dose levels. Dose-expansion: 30 patients were enrolled; 1 patient with NSCLC achieved PR lasting >77 weeks. Seven of 10 patients with melanoma (70%) and 7 of 20 patients with NSCLC (35%) achieved SD: median (range) duration of SD was 18.9 (13.9–49.0) weeks for the melanoma cohort versus 24.1 (14.3–77.9+) weeks for the NSCLC cohort; DCR (NSCLC) was 40%. Grade 3–4 treatment-emergent AEs were reported in 28 (49.1%) patients versus 11 (36.7%) patients in dose-escalation and dose-expansion, respectively. There were no grade 5 AEs deemed attributable to treatment. Ivuxolimab area under the concentration–time curve increased in a dose-dependent manner at 0.3–3 mg/kg doses.ConclusionsIvuxolimab+utomilumab was found to be well tolerated and demonstrated preliminary antitumor activity in selected groups of patients.Trial registration numberNCT02315066.
- Published
- 2022
20. Ovarian cancer: new strategies and emerging targets for the treatment of patients with advanced disease
- Author
-
Bradley J. Monk, Amy Jackson-Fisher, Ira Jacobs, Rebecca C. Arend, and Jeffrey Chou
- Subjects
0301 basic medicine ,Cancer Research ,targeted agents ,Angiogenesis ,medicine.medical_treatment ,Poly ADP ribose polymerase ,Review ,Carcinoma, Ovarian Epithelial ,03 medical and health sciences ,angiogenesis ,0302 clinical medicine ,Ovarian cancer ,medicine ,Humans ,PARP inhibitors ,Immune Checkpoint Inhibitors ,Pharmacology ,Chemotherapy ,biology ,Kinase ,business.industry ,medicine.disease ,Immune checkpoint ,Clinical trial ,Wee1 ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,PD-1/PD-L1 immune checkpoint inhibitors ,Molecular Medicine ,Female ,business - Abstract
Recently approved therapies have contributed to a significant progress in the management of ovarian cancer; yet, more options are needed to further improve outcomes in patients with advanced disease. Here we review the rationale and ongoing clinical trials of novel combination strategies involving chemotherapy, poly ADP ribose polymerase, programmed death 1 (PD-1)/PD-ligand 1 immune checkpoint and/or vascular endothelial growth factor receptor inhibitors. Further, we discuss novel agents aimed at targets associated with ovarian cancer growth or progression that are emerging as potential new treatment approaches. Among them, agents targeted to folate receptor α, tissue factor, and protein kinase-mediated pathways (WEE1 kinase, phosphatidylinositol-3 kinase α, cell cycle checkpoint kinase 1/2, ATR kinase) are currently in clinical development as mono- or combination therapies. If successful, findings from these extensive development efforts may further transform treatment of patients with advanced ovarian cancer.
- Published
- 2021
21. Airway profile of bioactive lipids predicts early progression of lung disease in cystic fibrosis
- Author
-
Hamed Horati, Bob J. Scholte, Camilla Margaroli, M. Veltman, Jeffrey Chou, Matthew B. Kilgore, Marta Stolarczyk, Hettie M. Janssens, Harm A.M.W. Tiddens, Joshua D. Chandler, Limin Peng, Rabindra Tirouvanziam, Pediatrics, and Cell biology
- Subjects
Male ,Pulmonary and Respiratory Medicine ,Pathology ,medicine.medical_specialty ,Cystic Fibrosis ,Respiratory System ,medicine.disease_cause ,Cystic fibrosis ,Article ,Bronchoscopy ,medicine ,Humans ,Longitudinal Studies ,Inflammation ,Bronchiectasis ,Lung ,biology ,medicine.diagnostic_test ,business.industry ,Infant ,respiratory system ,Lipid Metabolism ,medicine.disease ,Cystic fibrosis transmembrane conductance regulator ,respiratory tract diseases ,Oxidative Stress ,Bronchoalveolar lavage ,medicine.anatomical_structure ,Child, Preschool ,Neutrophil elastase ,Myeloperoxidase ,Pediatrics, Perinatology and Child Health ,Disease Progression ,biology.protein ,Cytokines ,Intercellular Signaling Peptides and Proteins ,Female ,Tomography, X-Ray Computed ,business ,Bronchoalveolar Lavage Fluid ,Biomarkers ,Oxidative stress - Abstract
BACKGROUND: Previously, we showed that abnormal levels of bioactive lipids in bronchoalveolar lavage fluid (BALF) from infants with cystic fibrosis (CF) correlated with early structural lung damage. METHOD: To extend these studies, BALF bioactive lipid measurement by mass spectrometry and chest computed tomography (CT, combined with the sensitive PRAGMA-CF scoring method) were performed longitudinally at 2-year intervals in a new cohort of CF children (n=21, aged 1–5 yrs). RESULTS: PRAGMA-CF, neutrophil elastase activity, and myeloperoxidase correlated with BALF lysolipids and isoprostanes, markers of oxidative stress, as well as prostaglandin E2 and combined ceramide precursors (Spearman’s Rho >0.5; P
- Published
- 2020
22. Immunogenicity of immunomodulatory, antibody-based, oncology therapeutics
- Author
-
Timothy P. Hickling, Jeffrey Chou, Ira Jacobs, Jasmine Davda, Paul Declerck, Siwen Hu-Lieskovan, Shahram Salek-Ardakani, and Eugenia Kraynov
- Subjects
0301 basic medicine ,Cancer Research ,Programmed Cell Death 1 Receptor ,Cell ,Review ,Immunotherapy, Adoptive ,B7-H1 Antigen ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Neoplasms ,Antibodies, Bispecific ,Immunology and Allergy ,Receptors, Chimeric Antigen ,biology ,Incidence ,Immunogenicity ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Treatment Outcome ,medicine.anatomical_structure ,Immune System Diseases ,Oncology ,030220 oncology & carcinogenesis ,Molecular Medicine ,Antibody ,Immunosuppressive Agents ,Immunomodulatory ,medicine.drug_class ,Immunology ,Monoclonal antibody ,Neutralizing antibodies ,PD-1/PD-L1 ,lcsh:RC254-282 ,Lymphocyte Depletion ,03 medical and health sciences ,Immune system ,medicine ,Humans ,Pharmacology ,business.industry ,Cancer ,medicine.disease ,Chimeric antigen receptor ,ADA ,030104 developmental biology ,CTLA-4 ,biology.protein ,business - Abstract
The increasing use of multiple immunomodulatory (IMD) agents for cancer therapies (e.g. antibodies targeting immune checkpoints, bispecific antibodies, and chimeric antigen receptor [CAR]-T cells), is raising questions on their potential immunogenicity and effects on treatment. In this review, we outline the mechanisms of action (MOA) of approved, antibody-based IMD agents, potentially related to their immunogenicity, and discuss the reported incidence of anti-drug antibodies (ADA) as well as their clinical relevance in patients with cancer. In addition, we discuss the impact of the administration route and potential strategies to reduce the incidence of ADA and manage treated patients. Analysis of published reports indicated that the risk of immunogenicity did not appear to correlate with the MOA of anti-programmed death 1 (PD-1)/PD-ligand 1 monoclonal antibodies nor to substantially affect treatment with most of these agents in the majority of patients evaluated to date. Treatment with B-cell depleting agents appears associated with a low risk of immunogenicity. No significant difference in ADA incidence was found between the intravenous and subcutaneous administration routes for a panel of non-oncology IMD antibodies. Additionally, while the data suggest a higher likelihood of immunogenicity for antibodies with T-cell or antigen-presenting cell (APC) targets versus B-cell targets, it is possible to have targets expressed on APCs or T cells and still have a low incidence of immunogenicity. Electronic supplementary material The online version of this article (10.1186/s40425-019-0586-0) contains supplementary material, which is available to authorized users.
- Published
- 2019
23. Addressing resistance to immune checkpoint inhibitor therapy: an urgent unmet need
- Author
-
Jeffrey Chou, Siwen Hu-Lieskovan, Ira Jacobs, Li Liu, Melissa Lynne Johnson, and Gabriel G. Malouf
- Subjects
0301 basic medicine ,Anticancer immunity ,Cancer Research ,Immune checkpoint inhibitors ,Bioinformatics ,Patient response ,Unmet needs ,03 medical and health sciences ,0302 clinical medicine ,Antigens, Neoplasm ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Tumor Microenvironment ,Medicine ,Humans ,Clinical efficacy ,Immune Checkpoint Inhibitors ,Tumor microenvironment ,business.industry ,Mechanism (biology) ,General Medicine ,Clinical trial ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Mutation ,business ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction - Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of various cancers by reversing the immunosuppressive mechanisms employed by tumors to restore anticancer immunity. Although ICIs have demonstrated substantial clinical efficacy, patient response can vary in depth and duration, and many do not respond at all or eventually develop resistance. ICI resistance mechanisms can be tumor-intrinsic, related to the tumor microenvironment or patient-specific factors. Multiple resistance mechanisms may be present within one tumor subtype, or heterogeneity exists among patients with the same tumor type. Consequently, designing effective combination treatment strategies is challenging. This review will discuss ICI resistance mechanisms, and summarize findings from key preclinical and clinical trials of ICIs, to identify potential treatment strategies or pathways to overcome ICI resistance.
- Published
- 2021
24. Pharmacologic Properties and Preclinical Activity of Sasanlimab, A High-affinity Engineered Anti-Human PD-1 Antibody
- Author
-
Brent Kern, Yasmina Noubia Abdiche, Allison Xu, HoangKim Nguyen, Eugenia Kraynov, Christopher R. Kimberlin, Shahram Salek-Ardakani, Cris Kamperschroer, Sherman M. Chin, Amir A. Al-Khami, Arvind Rajpal, Joyce Chou, Jerry D. Clark, Christopher P. Dillon, Natalija Budimir, Jeffrey Chou, John C. Lin, Bart Jessen, and Sawsan Youssef
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cancer immunotherapy ,In vivo ,Cell Line, Tumor ,Superantigen ,medicine ,Animals ,Humans ,Immune Checkpoint Inhibitors ,biology ,Chemistry ,Mixed lymphocyte reaction ,Immune checkpoint ,030104 developmental biology ,Cytokine ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Cytokine secretion ,Antibody - Abstract
Development of antagonistic mAbs that specifically target the immune checkpoint receptor, programmed cell death protein-1 (PD-1), is of great interest for cancer immunotherapy. Here, we report the biophysical characteristics and nonclinical antagonistic activities of sasanlimab (PF-06801591), a humanized anti-PD-1 antibody of IgG4 isotype. We show that sasanlimab binds selectively and with similar high potency to human and cynomolgus monkey PD-1 receptor and blocks its interaction with PD-L1 and PD-L2, with no detectable Fc-dependent effector function. The binding of sasanlimab to human and cynomolgus PD-1 is associated with the formation of a stable complex, which is likely to be the main driver of this high-affinity interaction. In vitro, sasanlimab significantly augmented T-cell proliferation and cytokine production in mixed lymphocyte reaction and superantigen stimulation assays. In vivo, sasanlimab accelerated the incidence of GvHD by enhancing T-cell proliferation and cytokine secretion in a xenogeneic model of acute GvHD and halted the growth of MC-38 colon adenocarcinoma tumors in human PD-1 knock-in mice. Pharmacokinetic and toxicokinetic findings from cynomolgus monkey showed that sasanlimab was active and well-tolerated. Taken together, the data presented here support the clinical development of sasanlimab for the treatment of patients with advanced cancers as a single agent or in combination with other immunotherapies.
- Published
- 2020
25. BMI Is a Better Body Proportionality Measure than the Ponderal Index and Weight-for-Length for Preterm Infants
- Author
-
Wendy N Ballew, Shannon C. Grabich, Rebecca Cantrell, Reese H. Clark, Irene E. Olsen, Jeffrey Chou, M. Louise Lawson, and A Nicole Ferguson
- Subjects
business.industry ,Gestational age ,Uncorrelated ,Weight for length ,Correlation ,03 medical and health sciences ,0302 clinical medicine ,Current practice ,030225 pediatrics ,Pediatrics, Perinatology and Child Health ,Medicine ,030212 general & internal medicine ,business ,Developmental Biology ,Demography - Abstract
Background: Clinicians have observed preterm infants in the neonatal intensive care unit growing disproportionally; however, the only growth charts that have been available were from preterm infants born in the 1950s which utilized the ponderal index. Prior to creating the recently published BMI curves, we found only 1 reference justifying the use of the ponderal index. Objectives: To determine the best measure of body proportionality for assessing growth in US preterm infants. Methods: Using a dataset of 391,681 infants, we determined the body proportionality measure that was most correlated with weight and least correlated with length. We examined the sex-specific overall correlations and then stratified further by gestational age (GA). We then plotted the body proportionality measures versus length to visualize apparent discrepancies in the appropriate measure. Results: The overall correlations showed weight/length3 (ponderal index) was the best measure but stratification by GA indicated that BMI (weight/length2) was the best measure. This seeming inconsistency was due to negative correlations between ponderal index and length at each GA. BMI, on the other hand, had a correlation with length across GAs, but was uncorrelated with length within GAs. Both ponderal index and BMI were positively correlated with weight. Conclusions: BMI is the appropriate measure of body proportionality for preterm infants, contrary to current practice.
- Published
- 2017
26. Preliminary Safety, Efficacy, Pharmacokinetics, and Pharmacodynamics of Subcutaneously (SC) Administered PF-06863135, a B-Cell Maturation Antigen (BCMA)-CD3 Bispecific Antibody, in Patients with Relapsed/Refractory Multiple Myeloma (RRMM)
- Author
-
Michael Sebag, Moshe Yair Levy, Andrzej Jakubowiak, Alexander M. Lesokhin, Andrew P. Dalovisio, Yogesh Jethava, Chandrasekhar Udata, Melhem Solh, Cristina Gasparetto, Heike Krupka, Nizar J. Bahlis, Jeffrey Chou, Caitlin Costello, Michael R. Savona, Noopur Raje, Michael P. Chu, Cynthia Basu, and Suzanne Trudel
- Subjects
education.field_of_study ,medicine.medical_specialty ,Bispecific antibody ,business.industry ,Immunology ,Population ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Peripheral blood ,Internal medicine ,Relapsed refractory ,medicine ,In patient ,Current employment ,education ,business ,Objective response ,Multiple myeloma - Abstract
Introduction: PF-06863135 (PF-3135) is a full-length, humanized, bispecific monoclonal antibody (mAb, IgG2a) targeting BCMA, which is highly expressed on multiple myeloma cells, and CD3 expressed on T cells. Intravenous (IV) dosing of PF-3135 has been previously evaluated at 0.1 - 50 μg/kg weekly with preliminary evidence of anti-myeloma activity in RRMM, without reaching the maximum tolerated dose (MTD). SC dose escalation was subsequently initiated given the potential to reduce the maximum concentration (Cmax), which is believed to be associated with cytokine release syndrome (CRS) and inflammatory response, with the intent of achieving a more favorable therapeutic window. Methods: Patients with RRMM previously treated with immunomodulatory drugs, proteasome inhibitors, and anti-CD38 mAb received SC dosages of PF-3135 at 80, 130, 215, and 360 μg/kg weekly. A modified toxicity probability interval method was used for dose escalation, with dose-limiting toxicities (DLTs) monitored in the first cycle (21 days). AEs were graded by CTCAE criteria except CRS, which was graded by Lee et al. 2014 criteria. Responses were assessed at the end of each cycle by the International Myeloma Working Group (IMWG) criteria. Peripheral blood was evaluated for soluble BCMA, cytokines, and immunophenotyping. PF-3135 pharmacokinetics and immunogenicity were also characterized. Results: As of April 15, 2020, 18 patients had received PF-3135 SC at 80 (n=6), 130 (n=4), 215 (n=4) and 360 (n=4) μg/kg weekly. The median number of prior anticancer treatment regimens was 7; all patients had received prior daratumumab therapy; 4 (22%) patients had received prior BCMA-targeted antibody-drug conjugate or chimeric antigen receptor T-cell therapy. All patients completed the DLT-monitoring period and none were observed. At cut-off date, dose escalation was ongoing with MTD not reached. All patients experienced treatment-emergent AEs (TEAEs); the most common were CRS reported in 11 (61%) patients, anemia in 9 (50%), thrombocytopenia in 7 (39%), and injection site reaction and lymphopenia in 6 (33% each). Grade (G) 3-4 TEAEs were observed in 12 (67%) patients, including G3 anemia in 8 (44%), G3 thrombocytopenia, lymphopenia, neutropenia, hypoalbuminemia, and bone pain in 2 (11% each), G4 lymphopenia in 4 (22%), G4 thrombocytopenia in 3 (17%), and G4 neutropenia in 2 (11%) patients. G5 TEAEs occurring in 3 (17%) patients (80 μg/kg group) were considered not treatment-related (disease progression [n=2] and sepsis [n=1]). CRS, the most common treatment-related AE, was observed across all dose levels in 11 patients (61% overall; 50% at 80 and 130 µg/kg; 75% at 215 and 360 µg/kg). Of these, 9 (50%) patients experienced G1 CRS and 2 (11%) had G2 CRS. CRS began within the first 2 days of treatment, with a median duration of 2 days (range 0-4). Per IMWG criteria, the objective response rate was 33% in the overall SC-dosed population and 75% at the top 2 dose levels (215 and 360 μg/kg). Two patients each achieved a best response of partial response (PR), very good PR (VGPR), and stringent complete response (sCR). Seven patients had best response of stable disease. Exposure to PF-3135 increased with dose in an approximately dose-proportional manner. SC administration resulted in a lower dose-normalized Cmax relative to IV administration and a prolonged absorption phase, with median time to Cmax ranging from 3 to 7 days. Consistent with the observed CRS, cytokine increases were the highest following the first dose and diminished significantly at subsequent doses. Levels of free, soluble BCMA decreased with increasing doses and demonstrated sustained decreases throughout each dose interval. Conclusions: AEs were generally manageable; CRS reported in the majority of patients was grade 1-2. Responses were achieved with SC dosing of PF-3135 in 6 of 8 (75%) patients at the 2 highest dose levels evaluated. Compared to IV, SC dosing did modulate the Cmax, so higher doses could be administered without observing increased severity of CRS. SC dose escalation is ongoing and additional results, including biomarkers and data from higher dose cohorts, will be presented. Clinicaltrials.gov identifier: NCT03269136. Funding: Pfizer. Disclosures Lesokhin: Janssen: Research Funding; Genentech: Research Funding; Takeda: Consultancy, Honoraria; Juno: Consultancy, Honoraria; GenMab: Consultancy, Honoraria; BMS: Consultancy, Honoraria, Research Funding; Serametrix Inc.: Patents & Royalties. Levy:Baylor University Med Center: Current Employment; Seattle Genetics: Consultancy, Honoraria, Research Funding; Sanofi: Consultancy, Honoraria, Research Funding; Takeda: Consultancy, Honoraria, Research Funding; Karyopharm: Consultancy, Honoraria, Research Funding; Janssen: Consultancy, Honoraria, Research Funding; Bristol Meyers Squibb: Consultancy, Honoraria, Research Funding; AstraZeneca: Consultancy, Honoraria, Research Funding; Amgen: Consultancy, Honoraria, Research Funding; AbbVie: Consultancy, Honoraria, Research Funding; BeiGene: Consultancy, Research Funding, Speakers Bureau. Bahlis:Sanofi: Consultancy, Honoraria; BMS/Celgene and Janssen: Consultancy, Honoraria, Other: Travel, Accomodations, Research Funding; Takeda: Consultancy, Honoraria; Amgen: Consultancy, Honoraria; Karyopharm Therapeutics: Consultancy, Honoraria; GSK: Consultancy, Honoraria; Genentech: Consultancy, Honoraria; AbbVie: Consultancy, Honoraria. Solh:Amgen: Consultancy, Speakers Bureau; BMS: Consultancy, Speakers Bureau; Seattle Genetics: Consultancy, Speakers Bureau; Partner Therapeutics: Research Funding; Pfizer: Research Funding; Pharmacyclics: Research Funding. Sebag:Celgene: Honoraria; Takeda: Honoraria; Amgen: Honoraria; Janssen: Honoraria, Research Funding. Jakubowiak:AbbVie, Amgen, BMS/Celgene, GSK, Janssen, Karyopharm: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Adaptive, Juno: Consultancy, Honoraria. Costello:Takeda: Consultancy, Honoraria, Research Funding; Celgene: Honoraria, Research Funding; Poseida Therapeutics: Research Funding; Janssen: Research Funding. Chu:BMS: Consultancy, Honoraria, Research Funding; Amgen: Consultancy, Honoraria; AstraZeneca: Consultancy, Honoraria; Gilead: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Teva: Consultancy, Honoraria. Savona:Boehringer Ingelheim: Patents & Royalties; Ryvu: Consultancy; Astex: Consultancy, Research Funding; Incyte: Consultancy, Research Funding; Takeda: Consultancy, Research Funding; Sunesis: Consultancy, Research Funding; Karyopharm: Consultancy, Current equity holder in publicly-traded company; Gilead: Consultancy; AbbVie: Consultancy; BMS: Consultancy; TG Therapeutics: Consultancy, Research Funding. Trudel:Sanofi: Honoraria; Takeda: Honoraria; Amgen: Consultancy, Research Funding; BMS: Consultancy, Honoraria, Research Funding; Genentech: Research Funding; GSK: Consultancy, Honoraria, Research Funding; Janssen: Honoraria, Research Funding; Pfizer: Honoraria, Research Funding; AstraZeneca: Honoraria; Karyopharm: Honoraria. Chou:Pfizer: Current Employment, Current equity holder in publicly-traded company. Udata:Pfizer: Current equity holder in publicly-traded company, Ended employment in the past 24 months. Basu:Pfizer: Current Employment, Current equity holder in publicly-traded company. Krupka:Pfizer: Current Employment, Current equity holder in publicly-traded company. Raje:Immuneel: Membership on an entity's Board of Directors or advisory committees; Caribou: Membership on an entity's Board of Directors or advisory committees; BMS: Consultancy; Astrazeneca: Consultancy; Takeda: Consultancy; Bluebird, Bio: Consultancy, Research Funding; Karyopharm: Consultancy; Janssen: Consultancy; Celgene: Consultancy; Amgen: Consultancy. OffLabel Disclosure: BCMA/CD3 bi-specific antibody in development for the treatment of patients with relapsed/refractory multiple myeloma
- Published
- 2020
27. Humanistic and economic impact of subcutaneous versus intravenous administration of oncology biologics
- Author
-
Rebecca C. Arend, Ira Jacobs, Ola Landgren, Jeffrey Chou, and Kenneth C. Anderson
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Cost-Benefit Analysis ,Injections, Subcutaneous ,Antineoplastic Agents ,03 medical and health sciences ,0302 clinical medicine ,Quality of life (healthcare) ,Internal medicine ,Neoplasms ,Health care ,medicine ,Humans ,Economic impact analysis ,Biological Products ,business.industry ,General Medicine ,Prognosis ,Cost savings ,030104 developmental biology ,Tolerability ,030220 oncology & carcinogenesis ,Administration, Intravenous ,business ,Healthcare providers ,Administration (government) ,Resource utilization - Abstract
More oncology biologics are becoming available for subcutaneous (sc.) administration and are expected to provide useful therapeutic options. We evaluated evidence published in the past 5 years to assess the humanistic and economic impact of sc. versus intravenous administration of approved cancer therapies and identify outcomes favoring either administration route. These publications focused predominantly on healthcare resource utilization and economic outcomes, demonstrating resource and cost savings with sc. administration. Patients reported a better health-related quality of life and preference for sc. formulations. Time-and-motion study analyses confirmed the convenience of sc. administration. These findings suggest that future availability of sc. oncology biologics, especially anti-PD-1/PD-ligand 1 antibodies due to their increased utility in various malignancies, may be beneficial for patients, healthcare providers and payers.
- Published
- 2019
28. Talimogene Laherparepvec in Combination With Ipilimumab in Previously Untreated, Unresectable Stage IIIB-IV Melanoma
- Author
-
Howard L. Kaufman, Kevin S. Gorski, Mohammed M. Milhem, David R. Minor, Jeffrey Chou, Robert H.I. Andtbacka, Ai Li, Igor Puzanov, Michael Chastain, Omid Hamid, Abraham Antonio Anderson, and Lisa Chen
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Skin Neoplasms ,Ipilimumab ,Gastroenterology ,03 medical and health sciences ,0302 clinical medicine ,T-Lymphocyte Subsets ,Internal medicine ,Original Reports ,medicine ,Clinical endpoint ,Humans ,Melanoma ,Survival analysis ,Aged ,Aged, 80 and over ,Oncolytic Virotherapy ,business.industry ,Antibodies, Monoclonal ,Middle Aged ,Stage iiib ,medicine.disease ,Tumor Burden ,Surgery ,Oncolytic virus ,Clinical trial ,Oncolytic Viruses ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Female ,Talimogene laherparepvec ,business ,medicine.drug - Abstract
Purpose Combining immunotherapeutic agents with different mechanisms of action may enhance efficacy. We describe the safety and efficacy of talimogene laherparepvec (T-VEC; an oncolytic virus) in combination with ipilimumab (a cytotoxic T-lymphocyte–associated antigen 4 checkpoint inhibitor) in patients with advanced melanoma. Methods In this open-label, multicenter, phase Ib trial of T-VEC in combination with ipilimumab, T-VEC was administered intratumorally in week 1 (106 plaque-forming units/mL), then in week 4 and every 2 weeks thereafter (108 plaque-forming units/mL). Ipilimumab (3 mg/kg) was administered intravenously every 3 weeks for four infusions, beginning in week 6. The primary end point was incidence of dose-limiting toxicities. Secondary end points were objective response rate by immune-related response criteria and safety. Results Median duration of treatment with T-VEC was 13.3 weeks (range, 2.0 to 95.4 weeks). Median follow-up time for survival analysis was 20.0 months (1.0 to 25.4 months). Nineteen patients were included in the safety analysis. No dose-limiting toxicities occurred, and no new safety signals were detected. Grade 3/4 treatment-related adverse events (AEs) were seen in 26.3% of patients; 15.8% had AEs attributed to T-VEC, and 21.1% had AEs attributed to ipilimumab. The objective response rate was 50%, and 44% of patients had a durable response lasting ≥ 6 months. Eighteen-month progression-free survival was 50%; 18-month overall survival was 67%. Conclusion T-VEC with ipilimumab had a tolerable safety profile, and the combination appeared to have greater efficacy than either T-VEC or ipilimumab monotherapy.
- Published
- 2016
29. Abstract CT138: Molecular markers in a phase I dose expansion study of PF-06801591 in locally advanced or metastatic non-small-cell lung cancer and urothelial carcinoma
- Author
-
Keith A. Ching, Xinmeng Jasmine Mu, Ira Jacobs, Melissa Lynne Johnson, Vinicius Bonato, Igor Bondarenko, Byoung Chul Cho, Joanna Pikiel, Hee Kyung Ahn, Alison Forgie, Jeffrey Chou, and Konstantin Penkov
- Subjects
Cancer Research ,business.industry ,medicine.drug_class ,Locally advanced ,Cancer ,Best Overall Response ,medicine.disease ,Monoclonal antibody ,Oncology ,Cancer research ,Medicine ,In patient ,Non small cell ,business ,Lung cancer ,Urothelial carcinoma - Abstract
Introduction: PF-06801591 is a humanized IgG4 monoclonal antibody that binds to the programmed cell death (PD-1) receptor and blocks its interaction with PD-1/2 ligands (PD-L1/2). In a phase 1 dose expansion study (NCT02573259), 106 patients with locally advanced or metastatic non-small cell lung cancer (NSCLC; n=68) or urothelial carcinoma (UC; n=38) were treated with PF-06891591 300 mg subcutaneously monthly. 12 patients with NSCLC (17.9%) and 7 with UC (18.4%) had a best overall response of partial response (PR). Response rates were higher in patients with high (≥1%) vs low ( Citation Format: Byoung Chul Cho, Konstantin Penkov, Igor Bondarenko, Joanna Pikiel, Hee Kyung Ahn, Alison Forgie, Ira Jacobs, Vinicius Bonato, Xinmeng Jasmine Mu, Keith Ching, Jeffrey Chou, Melissa L. Johnson. Molecular markers in a phase I dose expansion study of PF-06801591 in locally advanced or metastatic non-small-cell lung cancer and urothelial carcinoma [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr CT138.
- Published
- 2020
30. Abstract 4557: BCMA-CD3 bispecific antibody PF-06863135: Preclinical rationale for therapeutic combinations
- Author
-
Viola Lam, Puja Sapra, Ivana Djuretic, Chao-Pei Betty Chang, Andrea T. Hooper, Jeffrey Chou, Javier Chaparro-Riggers, Katarzyna Karwacz, and Heike Krupka
- Subjects
Cancer Research ,business.industry ,T cell ,medicine.disease ,Immune system ,medicine.anatomical_structure ,Oncology ,In vivo ,Blocking antibody ,medicine ,Cancer research ,Cytotoxic T cell ,business ,Multiple myeloma ,Ex vivo ,Lenalidomide ,medicine.drug - Abstract
BCMA-CD3 bispecific antibody PF-06863135 is currently under evaluation in an ongoing Phase 1 clinical trial in relapsed/refractory (RR) multiple myeloma (MM) patients (NCT03269136). We showed previously that BCMA-CD3 bispecific has potent single agent activity in RR MM human bone marrow aspirates in vitro and in preclinical models of human myeloma (Panowski et al, MCT 2019). Herein we used in vitro assays, ex vivo MM patient 3D cultures, and in vivo efficacy models to study therapeutic combinations of PF-06863135 with immune checkpoint inhibitors, immunomodulatory drugs (IMiD), and gamma secretase inhibitors (GSI). Upregulation of immune checkpoints, such as programmed cell death protein-1 (PD-1) or programmed death ligand 1 (PD-L1) has been demonstrated downstream of response to CD3 bispecifics. While PF-06863135 is effective as a single agent in ex vivo 3D primary MM cultures (EC50 0.2 nM), PD-L1 was upregulated on myeloma cells. We combined PF-06863135 with anti-PD-1 in subcutaneous and orthotopic myeloma models MM.1S and MM.1S-PD-L1 in NSG mice engrafted with human T cells. PF-06863135 demonstrated single agent anti-tumor activity in the MM.1S models; however, in the MM.1S-PD-L1 model, PF-06863135 activity was blunted. When given in combination with an anti-PD-1 blocking antibody, the full thrust of the single agent anti-tumor activity of PF-06863135 was restored. Dysfunctional, senescent like T cells may emerge downstream of T cell immunotherapies. IMiDs, such as lenalidomide, are a standard of care therapy in MM and result in immunomodulatory effects on T cells leading us to hypothesize PF-06863135 mediated tumor growth inhibition could be enhanced by combining with lenalidomide. When PF-06863135 and lenalidomide were given in combination in the human T cell engrafted established orthotopic MM.1S and MOLP8 models, an even greater anti-tumor activity was observed as compared to either agent alone. These studies suggest that PF-06863135 activity can be augmented by combinations with immunomodulatory agents such as anti-PD-1 or lenalidomide. BCMA is shed from the surface of myeloma cells by gamma secretases, not only reducing cell surface target density but contributing to a soluble sink. When treated with GSI in vitro, myeloma cell surface BCMA expression increased concurrent with reduction in soluble/shed BCMA across a panel of myeloma and lymphoma cell lines expressing a range of BCMA. In cytotoxic T lymphocyte co-culture assays, the activity of PF-06863135 was potentiated by combining with GSI. Studies of the combination of PF-06863135 and GSI are ongoing in preclinical in vivo models of myeloma. Taken together, our preclinical studies provide insights into mechanisms of action and resistance for PF-06863135, which has potential for profound single agent activity that can be enhanced with therapeutic combinations. Citation Format: Katarzyna Karwacz, Andrea T. Hooper, Chao-Pei Betty Chang, Heike Krupka, Jeffrey Chou, Viola Lam, Ivana Djuretic, Javier Chaparro-Riggers, Puja Sapra. BCMA-CD3 bispecific antibody PF-06863135: Preclinical rationale for therapeutic combinations [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 4557.
- Published
- 2020
31. Analysis of OX40 agonist antibody (PF-04518600) in patients with hepatocellular carcinoma
- Author
-
Andrea J. Bullock, Anthony B. El-Khoueiry, Omid Hamid, Wenjing Yang, Ferry A.L.M. Eskens, Carrie T. Taylor, Ruifeng Li, Jeffrey Chou, Alison Forgie, Jean-Philippe Spano, William P. Harris, Toshihiko Doi, Paul Gougis, Eric Angevin, and Ken Liao
- Subjects
Agonist ,Cancer Research ,biology ,medicine.drug_class ,business.industry ,SUPERFAMILY ,Monoclonal antibody ,medicine.disease ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,medicine ,biology.protein ,Cancer research ,Tumor necrosis factor alpha ,In patient ,Antibody ,Receptor ,business ,030215 immunology - Abstract
523 Background: PF-04518600 (PF-8600) is a humanized agonist IgG2 monoclonal antibody to the tumor necrosis factor superfamily receptor OX40. PF-8600 was given to patients (pts) with advanced/metastatic hepatocellular carcinoma (HCC) in dose expansion of a phase 1 study (NCT02315066). Safety and tolerability were primary endpoints and exploratory endpoints included biomarker analyses. Methods: Pts received either 30 mg (Arm 1) or 250 mg (Arm 2) of PF-8600 intravenously Q2W. Pts had pathologic diagnosis of advanced HCC, a Child-Pugh score of A or B7, and had ≤2 prior lines of therapy, or if treatment naïve, had declined standard of care. Radiological tumor assessments were conducted Q8W. Biopsy samples collected at baseline and wk 6 were analyzed by immunohistochemistry and RNA sequencing for pharmacodynamic (PD) analyses. Whole blood samples were collected longitudinally for DNA extraction for high-throughput sequencing of the T cell receptor β-chain. Results: Arm 1 enrolled 16 pts (mean age 65.6 yrs; range 54-81 yrs; prior PD-1/PD-L1: 5 pts) and Arm 2 enrolled 19 pts (mean age 61.7 yrs; range 26-79 yrs; prior PD-1/PD-L1: 3 pts). All grade treatment related adverse events (TRAEs) occurred in 69% of pts in Arm 1 and 58% of pts in Arm 2. The rate of ≥Grade 3 TRAEs was 31% and 16% in Arms 1 and 2, respectively. For both arms combined, the most common (≥10%) TRAEs were rashes and pruritus. In Arm 1, 8 pts (50%) and in Arm 2, 10 pts (53%) achieved stable disease (SD), with a mean duration of 18.4 (range: 14.0-30.3 wks) and 17.4 wks (range: 8.0-31.9 wks), respectively. PD effects were more evident in Arm 1 than Arm 2, including increased OX40 tumor expression and positive changes in gene signatures, reflecting an active anti-tumor immune response. Conclusions: PF-8600 was generally well tolerated and provided meaningful disease control. While safety and efficacy were not significantly different between the 2 doses, there were potential differences in the PD data. The safety and relative durability of SD in HCC pts may provide a rationale for exploration of combination therapy in this pt population. Clinical trial information: NCT02315066.
- Published
- 2020
32. Safety and clinical activity of subcutaneously (SC) administered anti-PD-1 antibody PF-06801591 in phase I dose-expansion cohorts of locally advanced or metastatic non-small cell lung cancer (NSCLC) and urothelial carcinoma (UC)
- Author
-
Y. Kulyaba, A. Kurochkin, K. Penkov, Alison Forgie, Jasmine Davda, Byoung Chul Cho, J. Pikiel, Hee Kyung Ahn, Igor Bondarenko, S. Odintsova, Ira Jacobs, Melissa Lynne Johnson, M. Korozan, Farhad Kazazi, Jeffrey Chou, Xiao Wang, and Ruifeng Li
- Subjects
medicine.medical_specialty ,business.industry ,Anti pd 1 ,Locally advanced ,Hematology ,Champions Oncology ,Phase i study ,Oncology ,Partial response ,Family medicine ,Injection site ,medicine ,In patient ,business ,Urothelial carcinoma - Abstract
Background The first-in-human, ongoing phase I study of PF-06801591, a humanized IgG4 antibody to programmed cell death 1 (PD-1) receptor, consisted of a Part 1 dose escalation and Part 2 dose expansion. In Part 1, PF-06801591 was administered intravenously or SC with an acceptable safety profile and antitumor activity in several solid tumor types. Herein, we report on Part 2 where we further evaluated SC PF-06801591 in patients (pts) with NSCLC and UC. Methods Anti-PD-1/L1-naive pts with NSCLC (n = 68) and UC (n = 38) received SC PF-06801591 300 mg q4w. NSCLC pts had ≤1 prior line of chemotherapy (CT) ± ≥1 ALK/EGFR mutation-directed therapies (if applicable) and UC pts had ≤2 lines of prior CT. Tumors were evaluated q8w by RECIST 1.1. Blood samples were collected for pharmacokinetic (PK) and immunogenicity assessments. Archival or baseline biopsies were evaluated by immunohistochemistry for PD-L1, RNA-seq, and whole exome sequencing. Results As of 27Dec2018, with median duration of treatment of 2.8 mos. in all 106 treated pts (55 still receiving treatment), 50.9% had treatment-related AEs (TRAEs) with 8.5% grade 3 or higher. Most common TRAEs were hyperthyroidism (9.4%), pruritus (6.6%), and increased amylase/lipase, anemia, hypothyroidism, pneumonitis, and rash (4.7% each). Treatment-emergent anti-drug antibodies were detected in 3 pts. With median follow-up of 5.1 mos. in 67 modified intent to treat (mITT) NSCLC pts, 11 (16.4%) pts achieved a partial response (PR) and 26 (38.8%) had stable disease (SD); the objective response rate (ORR) was 16.4% with 95% confidence interval (CI) 8.5-27.5% (25% in pts with ≥1% tumor cell PD-L1 expression). With median follow-up of 3.0 mos. in 37 mITT UC pts, 6 (16.2%) pts had a PR and 13 (35.1%) had SD; ORR was 16.2% (95% CI 6.2-32.0%). PK and biomarker data will be presented. Conclusions SC PF-06801591 given monthly was well tolerated, with minimal rates of injection site reactions and low incidence of immunogenicity. SC PF-06801591 demonstrated antitumor activity in NSCLC and UC pts at an early time point. The study is ongoing to further assess the durability of responses and long-term safety profile. Clinical trial identification NCT02573259. Editorial acknowledgement S. Mariani MD PhD of Engage Scientific Solutions, Southport, CT, funded by Pfizer and L. Chen. Legal entity responsible for the study Pfizer Inc. Funding Pfizer Inc. Disclosure B.C. Cho: Advisory / Consultancy, Research grant / Funding (institution): Novartis; Advisory / Consultancy, Research grant / Funding (institution): AstraZeneca; Research grant / Funding (institution): Bayer; Research grant / Funding (institution): MOGAM Institute; Research grant / Funding (institution): Dong-A ST; Research grant / Funding (institution), Licensing / Royalties: Champions Oncology; Advisory / Consultancy, Research grant / Funding (institution): Janssen; Advisory / Consultancy, Research grant / Funding (institution): Yuhan; Advisory / Consultancy, Research grant / Funding (institution): Ono; Research grant / Funding (institution): Dizal Pharma; Advisory / Consultancy, Research grant / Funding (institution): MSD; Advisory / Consultancy: Boehringer Ingelheim; Advisory / Consultancy: Roche; Advisory / Consultancy: BMS; Advisory / Consultancy: Pfizer; Advisory / Consultancy: Eli Lilly; Advisory / Consultancy: Takeda; Shareholder / Stockholder / Stock options: TheraCanVac Inc; Shareholder / Stockholder / Stock options: Gencurix Inc; Shareholder / Stockholder / Stock options: Bridgebio Therapeutics. K. Penkov: Research grant / Funding (institution): AbbVie; Research grant / Funding (institution): AstraZeneca; Research grant / Funding (institution): MSD; Research grant / Funding (institution): Pfizer; Research grant / Funding (institution): Prestige Biopharma; Research grant / Funding (institution): Regeneron; Research grant / Funding (institution): Tanvex. M. Korozan: Research grant / Funding (institution): AstraZeneca; Research grant / Funding (institution): Genentech/Roche; Research grant / Funding (institution): Regeneron. Y. Kulyaba: Research grant / Funding (institution): AstraZeneca; Research grant / Funding (institution): Azaya Therapeutics; Research grant / Funding (institution): Celltrion; Research grant / Funding (institution): GlaxoSmithKline; Research grant / Funding (institution): Janssen; Research grant / Funding (institution): Merck; Research grant / Funding (institution): Novartis; Research grant / Funding (institution): Pfizer; Research grant / Funding (institution): Regeneron; Research grant / Funding (institution): Roche; Research grant / Funding (institution): Synta Pharmaceuticals; Research grant / Funding (institution): Tanvex. H.K. Ahn: Honoraria (self): AstraZeneca; Honoraria (self): Boehringer Ingelheim; Honoraria (self): Eisai; Honoraria (self): Ono; Honoraria (self): Roche. S. Odintsova: Full / Part-time employment: Complete Medical Technologies. J. Davda: Shareholder / Stockholder / Stock options, Full / Part-time employment: Pfizer. A. Forgie: Shareholder / Stockholder / Stock options, Full / Part-time employment: Pfizer. X. Wang: Shareholder / Stockholder / Stock options, Full / Part-time employment: Pfizer. R. Li: Shareholder / Stockholder / Stock options, Full / Part-time employment: Pfizer. I.A. Jacobs: Shareholder / Stockholder / Stock options, Full / Part-time employment: Pfizer. F. Kazazi: Shareholder / Stockholder / Stock options, Full / Part-time employment: Pfizer. J. Chou: Shareholder / Stockholder / Stock options, Full / Part-time employment: Pfizer. M.L. Johnson: Honoraria (institution), Research grant / Funding (institution): AstraZeneca; Honoraria (institution), Research grant / Funding (institution): Genentech/Roche; Honoraria (institution), Research grant / Funding (institution): Boehringer Ingelheim; Honoraria (institution), Research grant / Funding (institution): Loxo; Honoraria (institution), Research grant / Funding (institution): Merck; Honoraria (institution), Research grant / Funding (institution): Mirati; Honoraria (institution), Research grant / Funding (institution): Sanofi; Honoraria (institution): Calithera; Honoraria (institution): Celgene; Research grant / Funding (institution): Lilly; Research grant / Funding (institution): EMD Serono; Research grant / Funding (institution): Janssen; Research grant / Funding (institution): GenMab; Research grant / Funding (institution): Pfizer; Research grant / Funding (institution): AbbVie; Research grant / Funding (institution): StemCentrix; Research grant / Funding (institution): Novartis; Research grant / Funding (institution): Incyte; Research grant / Funding (institution): Array BioPharma; Research grant / Funding (institution): Regeneron; Research grant / Funding (institution): Adaptimmune; Research grant / Funding (institution): Amgen; Research grant / Funding (institution): Apexigen; Research grant / Funding (institution): BeiGene; Research grant / Funding (institution): BerGenBio; Research grant / Funding (institution): Birdie; Research grant / Funding (institution): Checkpoint Therapeutics; Research grant / Funding (institution): Corvus; Research grant / Funding (institution): Cytomx; Research grant / Funding (institution): Daiichi–Sankyo; Research grant / Funding (institution): Dynavax; Research grant / Funding (institution): G1 Therapeutics; Research grant / Funding (institution): Genocea; Research grant / Funding (institution): Gritstone; Research grant / Funding (institution): Hengrui Therapeutics; Research grant / Funding (institution): Lycera; Research grant / Funding (institution): Neovia; Research grant / Funding (institution): Syndax; Research grant / Funding (institution): Tarveda; Spouse / Financial dependant, contract lobbyist : Astellas; Spouse / Financial dependant, contract lobbyist : Otsuka Pharmaceuticals. All other authors have declared no conflicts of interest.
- Published
- 2019
33. Tracking the Fate and Origin of Clinically Relevant Adoptively Transferred CD8+ T Cells In Vivo
- Author
-
Ryan O. Emerson, Kendall C. Shibuya, David G. Coffey, Harlan Robins, Felecia Wagener, Philip D. Greenberg, Edus H. Warren, Cindy Desmarais, Jeffrey Chou, Cassian Yee, Ivy Lai, Thomas M. Schmitt, Ilana M. Roberts, and Aude G. Chapuis
- Subjects
0301 basic medicine ,education.field_of_study ,Adoptive cell transfer ,Immunology ,T-cell receptor ,Population ,General Medicine ,Biology ,Article ,03 medical and health sciences ,CTL ,030104 developmental biology ,Antigen ,In vivo ,Monoclonal ,Cytotoxic T cell ,education - Abstract
Adoptively transferred tumor-specific cells can mediate tumor regression in cancers refractory to conventional therapy. Autologous polyclonal tumor-specific cytotoxic T cells (CTL) generated from peripheral blood and infused into patients with metastatic melanoma show enhanced persistence, compared to equivalent numbers of more extensively expanded monoclonal CTL, and are associated with complete remissions (CR) in select patients. We applied high-throughput T cell receptor Vβ sequencing (HTTCS) to identify individual clonotypes within CTL products, track them in vivo post-infusion and then deduce the pre-adoptive transfer (endogenous) frequencies of cells ultimately responsible for tumor regression. The summed in vivo post-transfer frequencies of the top 25 HTTCS-defined clonotypes originally detected in the infused CTL population were comparable to enumeration by binding of antigen peptide-HLA multimers, revealing quantitative HTTCS is a reliable, multimer-independent alternative. Surprisingly, the polyclonal CTL products were composed predominantly of clonotypes that were of very low frequency (VLF) in the endogenous samples, often below the limit of HTTCS detection (0.001%). In patients who achieved durable CRs, the composition of transferred CTLs was dominated (57-90%) by cells derived from a single VLF clonotype. Thus, HTTCS now reveals that tumor-specific CTL enabling long-term tumor control originate from endogenous VLF populations that exhibit proliferative/survival advantages. Along with results indicating that naive cell populations are most likely to contain cells that exist at VLF within the repertoire, our results provide a strong rationale for favoring T cells arising from VLF populations and with early-differentiation phenotypes when selecting subset populations for adoptive transfer.
- Published
- 2017
34. Safety, Clinical Activity, Pharmacokinetics, and Pharmacodynamics from a Phase I Study of PF-06863135, a B-Cell Maturation Antigen (BCMA)-CD3 Bispecific Antibody, in Patients with Relapsed/Refractory Multiple Myeloma (RRMM)
- Author
-
Alexander M. Lesokhin, Cristina Gasparetto, Heike Krupka, Jeffrey Chou, Cynthia Basu, Robert F. Cornell, Chandrasekhar Udata, Alison Forgie, Noopur Raje, Abraham C.F. Leung, Andrzej Jakubowiak, and Daniel Navarro
- Subjects
0301 basic medicine ,medicine.medical_specialty ,business.industry ,Immunology ,Cell Biology ,Hematology ,Neutropenia ,medicine.disease ,Biochemistry ,Phase i study ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Tolerability ,Leukocytopenia ,Pharmacokinetics ,Internal medicine ,Medicine ,In patient ,business ,Multiple myeloma ,Febrile neutropenia ,030215 immunology - Abstract
Introduction: PF-06863135 (PF-3135) is a bispecific, humanized, monoclonal antibody (mAb) consisting of BCMA- and CD3-targeting arms paired on an IgG2a backbone by hinge-mutation technology. PF-3135 binds BCMA+ myeloma cells and CD3+ T cells with affinities of 20 pM and ~40 nM, respectively (Panowski et al. Blood 2016). We report here findings from the dose-escalation portion of an ongoing, multi-center, open-label, phase I study (NCT03269136) of PF-3135 in patients with RRMM. Methods: Adult patients (≥18 years of age) with RRMM, previously treated with a proteasome inhibitor, an immunomodulatory drug, and an anti-CD38 mAb, received escalating, intravenous (IV) doses of PF-3135, once weekly. Prior BCMA-targeted bispecific T-cell engager or chimeric antigen receptor T-cell (CART) treatment was allowed by protocol. Patients had measurable disease per the International Myeloma Working Group (IMWG) updated criteria 2014. A modified toxicity probability interval method (mTPI), targeting a dose-limiting toxicity (DLT) rate of 25% (equivalence interval ± 5%) was used for dose escalation. The primary study objectives are to assess PF-3135 safety and tolerability, to determine the maximum tolerated dose (MTD) and select the recommended phase II dose (RP2D). Secondary objectives include evaluation of anti-myeloma activity, pharmacokinetics (PK), and immunogenicity of PF-3135. Results: As of April 9, 2019, 17 patients had received once weekly, non-continuous, IV infusion of PF-3135 in 6 dose-escalation groups. The majority were men (71%). The median age was 61 yrs (range, 47-82 yrs) and median disease duration since onset was 7 yrs (range, 1.1-13.3 yrs). Ten (59%) patients had ≥1 chromosomal abnormality and 5 (29%) had a normal karyotype (status not known for 2 [12%] patients). The median number of prior anti-myeloma therapies was 11; 5 (29%) patients had received prior BCMA-targeted therapy. Eight (47%) patients had relapsed MM and 8 (47%) had refractory disease (recurrence type not known for 1 [6%] patient). Ten (59%) patients experienced treatment-related (TR) AEs of any grade. Most TRAEs were grade 1-2, including cytokine release syndrome (CRS, 24%), thrombocytopenia (24%), anemia (18%), and pyrexia (18%). Three (18%) patients had grade 3 TRAEs (increased alanine aminotransferase/aspartate aminotransferase, leukocytopenia, neutropenia, and lymphopenia). One patient treated at the highest dose level, who had received prior BCMA CART therapy, developed treatment-related febrile neutropenia, a DLT, which may have been related to CRS and borderline/low neutrophil count at baseline. None of the patients had grade 4-5 TRAEs or discontinued treatment due to a TRAE. The median duration of treatment was 4 (range, 2-12) actual dosing days. Sixteen of the 17 patients were evaluable for response. At the time of data cut-off, one (6%) patient had a minimal response and 6 (35%) patients had stable disease (SD) across dose levels, as best response by investigator IMWG assessment; 9 (53%) patients experienced disease progression. The clinical benefit rate (defined as best response ≥SD) was 41% (95% CI: 18.4%, 67.1%). Conclusions: Treatment with IV PF-3135 was well tolerated at the dose levels evaluated. The observed CRS events were moderate and dose-dependent. Additional dose cohorts are accruing. The latest clinical, biomarker, and PK data will be presented for this ongoing study. Disclosures Raje: Medscape: Honoraria; Research to Practice: Honoraria; Takeda: Consultancy, Honoraria; Merck: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Celgene: Consultancy, Honoraria; BMS: Consultancy, Honoraria; Amgen: Consultancy, Honoraria; AstraZeneca: Research Funding. Jakubowiak:Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; GSK: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Takeda: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Adaptive Biotechnologies: Consultancy, Honoraria; Juno: Consultancy, Honoraria; Sanofi: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; SkyLineDx: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; AbbVie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; BMS: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; KaryoPharm Therapeutics: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Millennium: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees. Gasparetto:Janssen: Consultancy, Honoraria, Other: Travel, accommodations, or other expenses paid or reimbursed ; BMS: Consultancy, Honoraria, Other: Travel, accommodations, or other expenses paid or reimbursed ; Celgene: Consultancy, Honoraria, Other: Travel, accommodations, or other expenses paid or reimbursed . Cornell:KaryoPharm: Consultancy; Takeda: Consultancy. Krupka:Pfizer: Employment, Equity Ownership. Navarro:Pfizer: Employment, Equity Ownership. Forgie:Pfizer: Employment, Equity Ownership. Udata:Pfizer: Employment, Equity Ownership. Basu:Pfizer: Employment, Equity Ownership. Chou:Pfizer: Employment, Equity Ownership. Leung:Pfizer: Employment, Equity Ownership. Lesokhin:BMS: Consultancy, Honoraria, Research Funding; Serametrix Inc.: Patents & Royalties; Takeda: Consultancy, Honoraria; Genentech: Research Funding; Juno: Consultancy, Honoraria; GenMab: Consultancy, Honoraria; Janssen: Research Funding. OffLabel Disclosure: PF-06863135, investigational agent
- Published
- 2019
35. Abstract CT046: Predictive biomarkers and pharmacodynamic changes in tumor RNA expression in a phase I study of anti PD-1 monoclonal antibody PF-06801591
- Author
-
Vinicius Bonato, Fadi Braiteh, Siwen Hu-Lieskovan, Xiao Wang, Melissa Lynne Johnson, Juneko E. Grilley-Olson, Shobha Potluri, Alison Forgie, and Jeffrey Chou
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.diagnostic_test ,biology ,medicine.drug_class ,business.industry ,Cancer ,Cell cycle ,Monoclonal antibody ,medicine.disease ,Internal medicine ,Gene expression ,Biopsy ,medicine ,biology.protein ,Immunohistochemistry ,Antibody ,Receptor ,business - Abstract
Introduction: PF-06801591 is a humanized IgG4 monoclonal antibody that binds to programmed cell death (PD-1) receptor, blocking its interaction with PD-1 ligands. In a phase I study (NCT02573259; dose-escalation) of 40 patients (pts) with multiple solid tumor types treated with PF-06891591 intravenously (IV) or subcutaneously (SC), 7 pts showed partial response. PF-06801591 was well tolerated at all doses. Biopsy samples were evaluated for predictive biomarkers and pharmacodynamic effects. Methods: Biopsy samples at baseline and on-treatment (IV:day 29; SC:day 36) were collected from 4 dose cohorts (IV:1, 3 and 10 mg/kg; SC:300 mg). Whole-exome sequencing (WES) and RNA-seq of biopsy tissue were used to estimate tumor mutation burden (TMB) and gene expression. Regression analysis was used to identify TMB/genes/pathways potentially associated with response in baseline tissue. Differential analysis of baseline and on-treatment biopsies identified genes/pathways potentially upregulated by PF-06801591. PD-L1 (clone SP-263; Ventana) expression was evaluated in tumor biopsies by immunohistochemistry (IHC) and pathologist scoring. Results: In a combined WES analysis of samples from baseline biopsies (all doses, n=24 pts), higher TMB was significantly associated with improved objective response (R^2=0.215, P=0.013). From RNA-seq analysis, genes positively associated with response were involved in interferon-gamma (IFNG) and PD-1 signaling, and cell cycle; CTLA4 was among the genes most significantly associated with response (P Conclusion: Based on a mixed set of tumor types with possible sensitivity to anti-PD-1, high TMB, IFNG signaling, and PD-L1 were associated with response to PF-06801591, as reported for other anti-PD-1 antibodies. Albeit based on a small sample size and diverse indication profile, baseline expression of CTLA4 appears to be highly associated with response and PD-L1 RNA levels may be better associated with response than protein expression by IHC. Increased expression of genes/pathways associated with adaptive immune activation in on-treatment biopsies indicates an active, immunomodulatory mechanism with anti-PD-1 therapy. This ongoing study will evaluate predictive biomarkers in dose-expansion cohorts of PF-06801591 in non-small cell lung cancer and urothelial cancer. Acknowledgments Study sponsor: Pfizer. Medical writing support: Chu Kong Liew (Engage Scientific Solutions), funded by Pfizer. Citation Format: Siwen Hu-Lieskovan, Fadi Braiteh, Juneko E. Grilley-Olson, Shobha Potluri, Xiao Wang, Alison Forgie, Vinicius Bonato, Jeffrey Chou, Melissa L. Johnson. Predictive biomarkers and pharmacodynamic changes in tumor RNA expression in a phase I study of anti PD-1 monoclonal antibody PF-06801591 [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr CT046.
- Published
- 2019
36. Assessment of Subcutaneous vs Intravenous Administration of Anti–PD-1 Antibody PF-06801591 in Patients With Advanced Solid Tumors
- Author
-
Fadi Braiteh, Melissa Lynne Johnson, Ira Jacobs, Juneko E. Grilley-Olson, Siwen Hu-Lieskovan, Farhad Kazazi, Jasmine Davda, Jeffrey Chou, Ruifeng Li, and Alison Forgie
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,Injections, Subcutaneous ,Programmed Cell Death 1 Receptor ,Gastroenterology ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Pharmacokinetics ,Neoplasms ,Internal medicine ,Dose escalation ,Humans ,Medicine ,030212 general & internal medicine ,Dosing ,Adverse effect ,Aged ,biology ,business.industry ,Immunogenicity ,Antibodies, Monoclonal ,Middle Aged ,Antibodies, Neutralizing ,Clinical trial ,Treatment Outcome ,Oncology ,Immunoglobulin G ,030220 oncology & carcinogenesis ,Cohort ,biology.protein ,Administration, Intravenous ,Female ,Antibody ,business - Abstract
Importance We assessed feasibility of monthly subcutaneous administration of PF-06801591, a humanized immunoglobulin G4 monoclonal antibody that binds to the programmed cell death (PD-1) receptor and blocks its interaction with PD-1 ligands. Objective To evaluate the safety, efficacy, and pharmacokinetics of PF-06801591 administered intravenously vs subcutaneously. Design, Setting, and Participants Ongoing phase 1, open-label, multicenter, dose-escalation study of 40 patients, 18 years or older, with locally advanced or metastatic solid tumors, enrolled between March 8, 2016, and March 5, 2018, from 4 US medical centers. Interventions An intravenous dose of 0.5, 1, 3, or 10 mg/kg of PF-06801591 was administered every 3 weeks or a subcutaneous dose of 300 mg was administered every 4 weeks. Dose escalation occurred after 2 to 4 patients were enrolled per dose level, with additional patients enrolled in each cohort for further assessment. Main Outcomes and Measures The primary end points were dose-limiting toxic effects and safety. Secondary end points included pharmacokinetics, immunogenicity, PD-1 receptor occupancy, and efficacy. Results Of 40 enrolled patients (12 men and 28 women; mean [SD] age, 61 [13] years) in this phase 1 dose-escalation trial, 25 received PF-06801591 intravenously at escalating dose levels (0.5, 1, 3, or 10 mg/kg) and 15 patients received the monoclonal antibody subcutaneously at a single dose level. No dose-limiting toxic effects were observed. Grade 3 or higher treatment-related adverse events occurred in 4 (16%) patients treated intravenously and 1 (6.7%) patient treated subcutaneously. Immune-related adverse events occurred in 10 (40%) patients treated intravenously and 3 (20%) treated subcutaneously. No dose–adverse event associations were observed during intravenous dose escalation, and no serious skin toxic effects occurred with subcutaneous delivery. Responses were seen in 5 patients receiving PF-06801591 intravenously and in 2 patients treated subcutaneously for an overall objective response rate of 18.4%. Median overall survival was not reached with intravenous dosing vs 10.7 months with subcutaneous administration. Exposure to PF-06801591 increased in a dose-proportional manner over the range of intravenous doses. Median time to maximum observed serum concentration was 8 days after subcutaneous administration. Full PD-1 receptor occupancy was seen in all dose cohorts. Conclusions and Relevance Anti–PD-1 antibody PF-06801591 was tolerable and showed antitumor activity in a variety of tumor types across all dose levels of intravenous and subcutaneous administration. Monthly subcutaneous administration of PF-06801591 offers a convenient, effective alternative to currently available intravenously administered checkpoint inhibitors. Trial Registration ClinicalTrials.gov identifier:NCT02573259
- Published
- 2019
37. High-throughput sequencing of B- and T-lymphocyte antigen receptors in hematology
- Author
-
Edus H. Warren, Frederick A. Matsen, and Jeffrey Chou
- Subjects
Receptors, Antigen, T-Cell, alpha-beta ,T cell ,Immunology ,Population ,Receptors, Antigen, B-Cell ,Biology ,Crystallography, X-Ray ,Biochemistry ,Antigen ,medicine ,Humans ,Cytotoxic T cell ,Antigen-presenting cell ,education ,BLOOD Spotlight ,education.field_of_study ,T-cell receptor ,Hematopoietic Stem Cell Transplantation ,High-Throughput Nucleotide Sequencing ,Receptors, Antigen, T-Cell, gamma-delta ,Cell Biology ,Hematology ,Neoplastic Cells, Circulating ,Molecular biology ,Chimeric antigen receptor ,Protein Structure, Tertiary ,medicine.anatomical_structure ,Hematologic Neoplasms ,biology.protein ,Antibody ,Dimerization - Abstract
Application of high-throughput DNA sequencing to the analysis of B- and T-lymphocyte antigen receptors has great potential for improving the monitoring of lymphoid malignancies, assessing immune reconstitution after hematopoietic cell transplantation, and characterizing the composition of lymphocyte repertoires. Current technology can define the number and frequency of immunoglobulin heavy, T-cell receptor (TCR)α, TCRβ, or TCRγ chains expressed in a population of lymphocytes; techniques for determining the number of antigen receptor heterodimers, such as TCRαβ pairs, expressed in the population are under development.
- Published
- 2013
38. Origin and evolution of the T cell repertoire after posttransplantation cyclophosphamide
- Author
-
David G. Coffey, Andrea M. H. Towlerton, Leo Luznik, Christopher G. Kanakry, Harlan Robins, Jeffrey Chou, Barry E. Storer, Christopher D. Gocke, Edus H. Warren, Paul O'Donnell, Cecilia C.S. Yeung, and Ante Vulic
- Subjects
0301 basic medicine ,medicine.diagnostic_test ,Cyclophosphamide ,T cell ,Lymphocyte ,T-cell receptor ,General Medicine ,Biology ,Flow cytometry ,Transplantation ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,medicine.anatomical_structure ,Immunology ,medicine ,CD8 ,030215 immunology ,medicine.drug ,Research Article - Abstract
Posttransplantation cyclophosphamide (PTCy) effectively prevents graft-versus-host disease (GVHD), but its immunologic impact is poorly understood. We assessed lymphocyte reconstitution via flow cytometry (n = 74) and antigen receptor sequencing (n = 35) in recipients of myeloablative, HLA-matched allogeneic BM transplantation using PTCy. Recovering T cells were primarily phenotypically effector memory with lower T cell receptor β (TRB) repertoire diversity than input donor repertoires. Recovering B cells were predominantly naive with immunoglobulin heavy chain locus (IGH) repertoire diversity similar to donors. Numerical T cell reconstitution and TRB diversity were strongly associated with recipient cytomegalovirus seropositivity. Global similarity between input donor and recipient posttransplant repertoires was uniformly low at 1-2 months after transplant but increased over the balance of the first posttransplant year. Blood TRB repertoires at ≥3 months after transplant were often dominated by clones present in the donor blood/marrow memory CD8+ compartment. Limited overlap was observed between the TRB repertoires of T cells infiltrating the skin or gastrointestinal tract versus the blood. Although public TRB sequences associated with herpesvirus- or alloantigen-specific CD8+ T cells were detected in some patients, posttransplant TRB and IGH repertoires were unique to each individual. These data define the immune dynamics occurring after PTCy and establish a benchmark against which immune recovery after other transplantation approaches can be compared.
- Published
- 2016
39. Melanoma and immunotherapy bridge 2015 : Naples, Italy. 1-5 December 2015
- Author
-
Erik Wennerberg, Gabriele Madonna, Gennaro Ciliberto, Michele Minopoli, Caroline Dutriaux, Matthew Wongchenko, Elisabetta Gambale, David F. Stroncek, Céleste Lebbé, Ani S. Balmanoukian, Gianluca Di Monta, Vincenzo Ingangi, Soldano Ferrone, Ivan Marquez-Rodas, Giuseppe Masucci, Janis M. Taube, Simona Mastroeni, Gerardo Bott, Franck Pagès, Jonathan M. Pitt, Judit Olasz, Elisabetta Panza, Paola Michelozzi, Daniil Stoyakovskiy, Stéphane Dalle, Mario Sznol, John M. Kirkwood, Keith T. Flaherty, Maria Capuano, Amalia Azzariti, Edward Cha, Peter Boasberg, Maria Godeny, Angela Gismondi, Ornella Franzese, Giusy Gentilcore, Jean-Jacques Grob, Olivier Michielin, Adina Elena Stanciu, Giuseppe Cirino, Julien Fourcade, Nelofer Syed, Giuseppe Ercolano, Caroline Robert, Ascierto Paolo Antonio, Christine K. Gause, Silviu Voinea, Adeeb Rahman, Anne Caignard, Camila Flores, Cristina Fortes, Yuya Yoshimoto, Angela Sandru, Andras Szollar, Monica Cantile, Frederic Lehmann, Maria Libera Ascierto, Sacha Gnjatic, Marco Tucci, Rosa Russo, Giuseppina Liguori, Valeria De Biasio, David Ross Kaufman, Mary Ruisi, Ewa Kalinka-Warzocha, Phillip Wong, Rosaria Falcon, Vincenzo Faiola, Nicole Richie, Lars Ny, Miri Blank, Paola De Cicco, Anna Passarelli, Jean-François Baurain, Guido Kroemer, Claudio Jommi, Francesca Capone, Maria Teresa Fierro, Tracee L. McMiller, Lev V. Demidov, Alessandro Testori, Omid Hamid, Marone Ugo, Annamaria Anniciello, Andrew J. Park, Fara De Murtas, RuthAnn Gordan, Emil Farkas, David Hogg, Alessandra Di Paolo, Mark Maurer, Yangyang Wang, Mario Mandalà, Rodabe N. Amaria, Massimiliano Di Marzo, Stefania Guida, Luigi Fattore, Veronica Huber, Ludmila Danilova, Luigi Aurisicchio, Gabriella Aquino, Domenico Mallardo, Catriona M. McNeil, Stephanie Anne Kronenberg, Consiglia Carella, Theresa S. Pritchard, Katia Bifulco, Michaela Semeraro, Carlo M. Croce, David P. Enot, Laurence Zitvogel, Marcella Occelli, Benjamin Weide, Magdalena Thurin, Margherita Cerrone, Naiyer A. Rizvi, Blessing Agunwamba, Stella D'Oronzo, Sarah Jegou, Stucci Stefania, Drew M. Pardoll, Vito Michele Garrisi, Haidong Tang, Szabolcs Horvath, Hong Wang, Benjamin Brady, Antonio Doronzo, Claudia Marino, Xian He, Michael A. Davies, Hexiao Wang, Isabelle Rooney, Orsolya Csuka, Maurizio Nudo, Lance Leopold, Jeffrey S. Wasser, Sabino Strippoli, Silvia Ch Formenti, MariaLaura Foddai, Michael A. Postow, Robert H.I. Andtbacka, Paul Lorigan, Tommy Andersson, Naoko Imai, Ari VanderWalde, Mariaelena Capone, Ilsung Chang, Laura Lattanzio, Carmen Loquai, Arantxa Sancho, Christine Horak, Federica Sallusto, Timea Balatoni, Maha Ayyoub, Angela Santoni, Alessio Caggiati, Anna Lisa De Presbiteris, Henrik Schmidt, Paola Savoia, Pontus Berglund, Igor Puzanov, Aurélien Marabelle, Ana Carrizossa Anderson, Hassane M. Zarour, Maria Wolodarski, Patrick Hwu, Joel Jiang, Pio Zeppa, Jeffrey A. Sosman, Eugenio Fernandez, Susan Costantini, Marcus Ballinger, Luc Thomas, Leslie Cope, Rolf Kiessling, Christophe Borg, Francesca Platini, Florian Stratica, Tilman T. Rau, Craig L. Slingluff, Paolo A. Ascierto, Angela Ianaro, Harriet M. Kluger, Stephen Hodi, Tara C. Gangadhar, Claire Vanpouille-Box, Caroline Flament, Hearn J. Cho, Mannavola Francesco, Takahiro Yamazaki, Charles G. Drake, Jason J. Luke, Miklos Kasler, Linda Pan, Caracò Corrado, Alfonso Berrocal, Angel Rivera, Vera Hirsh, Eduardo J. Patriarca, Giovanni Di Giulio, Antonello Pessi, Helena Stabile, Helene Hardy, Tucci Marco, Ralph Venhaus, Maria Luisa Di Cecilia, Catherine A. Harwood, Jonathan Cebon, Anna Maria Anniciello, Grant A. McArthur, Carlo Baldi, Ahmad A. Tarhini, Shelley Coleman, Gil Bar-Sela, Axel Hauschild, Byoung S. Kwon, Maria Paula Roberti, Sabin Cinca, Tiziana Cocco, Valeria Sorrentino, Jeffrey Wallin, Rosa Camerlingo, Sandra Demaria, Jedd D. Wolchok, Isabel Poschke, Alessandro Lugli, Michael B. Atkins, Andrea Cavalcanti, Laura Marra, Rosamaria Pinto, Adam D. Cohen, Michele Maio, Weiyi Peng, Rosario Guarrasi, David Enot, Antoni Ribas, Oscar Alabiso, Chiara Armogida, Silvestris Franco, Jessica C. Hassel, Rita Mancini, Michele Guida, Silvia C. Formenti, Andrea P. Sponghini, Imma Maida, Alba Zappalà, Charlotte M. Proby, Alan J. Korman, Yibing Yan, Matias Chacon, Haiying Xu, Carolin Blattner, Maria-Paula Roberti, Lisa Chen, James Larkin, Ryan J. Sullivan, Madonna Gabriele, Nadege Vimond, Cosmo Di Donna, Farnaz Moradi, Manish R. Patel, Sylvie Rusakiewicz, Francesca Passarelli, Luis de la Cruz-Merino, Nicolas Jacquelot, Roberta Miceli, Viktor Umansky, Akos Savolt, David Rondonotti, Gabriella Liszkay, Jianda Yuan, Stefani Spranger, Yufan Zhao, Yehuda Shoenfeld, Todd M. Bauer, Claus Garbe, Joe-Marc Chauvin, Achim A. Jungbluth, Cristiana Lo Nigro, Alexander M. Lesokhin, Gabriella Guida, Brigitte Dréno, Cindy Sanders, Jeffrey S. Weber, Janet Maleski, Chris Cheadle, Romain Daillère, Isabella Sperduti, Michaele Maio, Claudia Felici, Parneet K. Cheema, Concetta Ragone, Johan Hansson, Klara Eles, Victoria Atkinson, Speiser Daniel, Daniel O. Koralek, Zhaojun Sun, Debora Malpicci, Elena Marra, Rickard Linnskog, Jeffrey Chou, Yang Wang, Eugenia Panaitescu, F. Stephen Hodi, Anthony J. Olszanski, Chiara Botti, Nicola Mozzillo, Anna Ferretta, Paul B. Chapman, Michaë la Semeraro, Belinda Palermo, Francesco Sabbatino, Neil H. Segal, Yago Pico de Coaña, Tseng-hui Timothy Chen, Ornella Pagliano, John B. A. G. Haanen, Huibin Yue, Emilia Caputo, Alan E. Berger, Simona De Summa, Nikoletta Lendvai, Paola Queirolo, Francesco Mannavola, Thomas F. Gajewski, Michele De Tursi, Paola Nisticò, Karen Briscoe, Karmele Mujika Eizmendi, Maria Vincenza Carrier, Passarelli Anna, Laurent Mortier, Crescenzo D'Alterio, Jorge Camarero, Licia Rivoltini, Pietro Quaglino, Davide Quaresmini, Marie Vétizou, Anna Maria Di Giacomo, Chandra Prakash Prasad, Riccardo Bono, Vichnou Poirier-Colame, David Smith, Capone Mariaelena, Giosuè Scognamiglio, Margaret K. Callahan, Vashisht Gopal Yennu Nanda, Fabiola Gilda Cordaro, George J. Netto, Madalina Bolovan, Federica Fratangelo, Josep Malvehy, Robert A. Anders, Karsten A. Pilones, Vincenzo C. Battarra, Karin Leandersson, Maria Letizia Motti, Yang Xin Fu, Tim Crook, Sarah Nataraj, Alastair M. Thompson, Franco Silvestris, Carolina Cauchi, Georgina V. Long, Holbrook E Kohrt, Giuseppe Pirozzi, Celeste Fusciello, Marco Carlo Merlano, François Aubin, Mozzillo Nicola, Giuseppe Cirin, Stefania Scala, Suzanne L. Topalian, Alexander M.M. Eggermont, Antonio Marra, Jinshui Fan, Reinhard Dummer, Federica Zito Marino, Amanda Ralabate, Matthew M. Burke, Piotr Rutkowski, Gerardo Botti, Stefano Pepe, Ivor Caro, and Stefania Tommasi
- Subjects
0301 basic medicine ,business.industry ,MEDLINE ,General Medicine ,medicine.disease ,Bridge (interpersonal) ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Medicine ,Medical emergency ,business - Published
- 2016
40. CD8+ T-cell Clones Specific for the 5T4 Antigen Target Renal Cell Carcinoma Tumor-initiating Cells in a Murine Xenograft Model
- Author
-
Richard Harrop, Janise D. Deming, Jeffrey Chou, Scott S. Tykodi, Edus H. Warren, and Shoko Satoh
- Subjects
Cytotoxicity, Immunologic ,Cancer Research ,medicine.medical_treatment ,Immunology ,Cell Separation ,Mice, SCID ,CD8-Positive T-Lymphocytes ,Biology ,Cancer Vaccines ,Immunotherapy, Adoptive ,Article ,Epitope ,Mice ,Antigen ,Cell Line, Tumor ,HLA-A2 Antigen ,medicine ,Animals ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,Carcinoma, Renal Cell ,Pharmacology ,Membrane Glycoproteins ,Immunotherapy ,Flow Cytometry ,Xenograft Model Antitumor Assays ,Molecular biology ,Kidney Neoplasms ,Peptide Fragments ,Tumor antigen ,Clone Cells ,CTL ,Clone (B-cell biology) ,CD8 ,Protein Binding - Abstract
The tumor antigen 5T4 is frequently expressed at high levels on renal cell carcinoma (RCC) and other epithelial carcinomas. Surveys of normal tissues demonstrate abundant 5T4 expression on placental trophoblast cells with limited expression elsewhere. 5T4 is the target for a therapeutic cancer vaccine (MVA-5T4) that elicits 5T4-specific serological, proliferative, and cytotoxic T lymphocyte (CTL) responses. However, the antitumor activity of 5T4-specific CTL has not been extensively characterized. CD8 T cells from HLA-A2 healthy donors (n=4) or RCC patients (n=2) were stimulated in vitro with the HLA-A2-binding nonamer peptides 5T417-25 or 5T497-105 and screened by flow cytometry with specific tetramers (TET). CD8/TET T-cell clones specific for 5T417-25 or 5T497-105 peptide were isolated from 4/6 and 1/4 donors, respectively. A subset of clones specific for 5T417-25 was cytolytic for MVA-5T4-infected HLA-A2 EBV-transformed lymphoblastoid cell line target cells and for constitutively HLA-A2-expressing and 5T4-expressing RCC tumor cell lines (including A498 RCC). In a xenoengraftment assay, the coinoculation of a representative 5T417-25-specific CTL clone with A498 RCC tumors cells into immune-deficient mice completely prevented growth of A498 tumors. Taken together, these data demonstrate high-avidity CD8 CTL able to recognize the naturally processed 5T417-25 epitope on RCC tumor cells including putative tumor-initiating cells are present in peripheral blood of both healthy donors and RCC patients. CD8T-cell immunity targeting 5T417-25 is therefore of substantial interest both as a potential target for further development of vaccination or adoptive cellular immunotherapy and for immune monitoring studies in association with nonspecific immunotherapies.
- Published
- 2012
41. Pharmacodynamic (PD) changes in tumors and peripheral blood T cell receptor (TCR) repertoire in a phase I study combining OX40 (PF-04518600) and 4-1BB (utomilumab) agonistic monoclonal antibodies (mAbs)
- Author
-
Adi Diab, Jeffrey S. Wasser, Patrick A. Ott, Shobha Potluri, Eric Angevin, Omid Hamid, J-P. Spano, John A. Thompson, Anthony B. El-Khoueiry, Alberto Chiappori, Heike Krupka, Toshihiko Doi, Naiyer A. Rizvi, Ferry A.L.M. Eskens, Tenshang Joh, Willeke Ros, Jeffrey Chou, Xiao Wang, B.J. Ganguli, and Siwen Hu-Lieskovan
- Subjects
medicine.drug_class ,business.industry ,T-cell receptor ,Hematology ,Tcr repertoire ,Monoclonal antibody ,Peripheral blood ,Phase i study ,Oncology ,Pharmacodynamics ,Immunology ,Agonistic behaviour ,medicine ,Utomilumab ,business - Published
- 2018
42. Abstract CT010: Pharmacodynamic (PD) changes in tumor RNA expression and the peripheral blood T cell receptor (TCR) repertoire in a phase I study of OX40 agonistic monoclonal antibody (mAb) PF-04518600 (PF-8600)
- Author
-
Siwen Hu-Lieskovan, Tenshang Joh, Toshihiko Doi, Omid Hamid, Jeffrey Chou, John A. Thompson, Willeke Ros, Catherine Fleener, Xiao Wang, Shoba Potluri, Adi Diab, Hua Long, Carrie T. Taylor, Anthony B. El-Khoueiry, Bishu J. Ganguli, and F. Eskens
- Subjects
0301 basic medicine ,Cancer Research ,Melanoma ,T cell ,T-cell receptor ,Biology ,medicine.disease ,Molecular biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,medicine.anatomical_structure ,Oncology ,Interferon ,030220 oncology & carcinogenesis ,medicine ,Cytotoxic T cell ,Utomilumab ,CD8 ,medicine.drug - Abstract
Introduction: PF-8600 is a fully human IgG2 agonistic mAb against the tumor necrosis factor superfamily receptor OX40. OX40 improves T cell survival, proliferation, and activation and may enhance anti-tumor immunity. In an ongoing phase I study (NCT02315066) of 52 patients (pts) with melanoma, hepatocellular carcinoma, head and neck carcinoma, or renal cell carcinoma treated with PF-8600 monotherapy in dose escalation, 2 pts had best overall response of partial response (PR) and 27 of stable disease. PF-8600 was well-tolerated at all doses. Peripheral blood flow cytometry had previously shown increased proliferation and activation of CD4 central memory T cells at certain dose levels, suggesting a PD effect. RNAseq analysis of tumor biopsy samples and TCR sequencing of peripheral blood were used to further support proof of mechanism. Methods: Biopsy samples at baseline and wk 6 were collected from 5 dose cohorts [0.1 (4), 0.3 (3), 1.5 (4), 3.0 (3) and 10.0 (2), dose in mg/kg (n)]. Biopsy tissue was analyzed by RNAseq and gene ranking-based gene set enrichment analysis (FGSEA) to identify immune pathways potentially up-regulated by OX40. CD4/CD8 cells were isolated from peripheral blood samples from 4 dose cohorts (0.1, 0.3, 1.5, and 3.0 mg/kg). DNA was extracted for sequencing of the TCR-β chain complementarity-determining region 3 (CDR3). Results: In a combined analysis of samples from pts dosed with ≥1.5 mg/kg, the top 3 gene sets showing enrichment at wk 6 of therapy were associated with inflammatory response, interferon-γ response and allograft rejection. These gene sets were identical to the top 3 most enriched in tumor from a syngeneic mouse tumor model exposed to a murine OX40 agonist. This pattern of enrichment was not observed if samples from lower doses were included in the analysis. TCR sequencing revealed clonal expansion of CD4/CD8 T cells at all dose levels at wk 6 [CD4: mean = 8 expanded clones per 100,000 clones (range = 1 - 80), CD8: mean = 56 (range = 1 - 500)]. The 2 patients with PR had among the lowest numbers of expanded CD4 and CD8 clones (CD4: 4 and 2; CD8: 7 and 4). Conclusion: Enrichment of gene sets associated with immune activation in tumor tissue from patients dosed with PF-8600 provides evidence supporting an active, immunomodulatory mechanism. Peripheral CD4/CD8 T cell populations exhibited clonal expansion in response to dosing with PF-8600 at all dose levels further suggesting a PD effect. However, clinical response did not necessarily correlate with a high number of expanded T cell clones, suggesting that clinical response to OX-40 agonism may be driven by the expansion of select anti-tumor T cell clones rather than a broad expansion of T cell clones in the peripheral blood. The phase I study will continue to evaluate PD changes in the tumor and peripheral blood in dose-expansion cohorts of PF-8600 ± utomilumab. Citation Format: Adi Diab, Omid Hamid, John A. Thompson, Willeke Ros, Fredericus A. L. M. Eskens, Toshihiko Doi, Siwen Hu-Lieskovan, Hua Long, Tenshang Joh, Shoba Potluri, Xiao Wang, Catherine Fleener, Carrie Turich Taylor, Bishu J. Ganguli, Jeffrey Chou, Anthony B. El-Khoueiry. Pharmacodynamic (PD) changes in tumor RNA expression and the peripheral blood T cell receptor (TCR) repertoire in a phase I study of OX40 agonistic monoclonal antibody (mAb) PF-04518600 (PF-8600) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr CT010.
- Published
- 2018
43. A Phase I/III, multicenter, open-label trial of talimogene laherparepvec (T-VEC) in combination with pembrolizumab for the treatment of unresected, stage IIIb-IV melanoma (MASTERKEY-265)
- Author
-
Olivier Michielin, Josep Malvehy, Olga M. Kuznetsova, Jeffrey Chou, F. Stephen Hodi, Anthony J. Olszanski, Ari M. Vanderwalde, Thomas F. Gajewski, John M. Kirkwood, Jonathan Cebon, David Ross Kaufman, Reinhard Dummer, Igor Puzanov, Eugenio Fernandez, Antoni Ribas, Robert H.I. Andtbacka, Georgina V. Long, and Lisa Chen
- Subjects
Pharmacology ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,viruses ,medicine.medical_treatment ,Melanoma ,Immunology ,Ipilimumab ,Immunotherapy ,Pembrolizumab ,medicine.disease ,Oncolytic virus ,Unresected ,Internal medicine ,Poster Presentation ,medicine ,Clinical endpoint ,Molecular Medicine ,Immunology and Allergy ,business ,Talimogene laherparepvec ,medicine.drug - Abstract
Meeting abstracts T-VEC is a herpes simplex virus-1-based oncolytic immunotherapy designed to selectively replicate in tumors, produce GM-CSF, and stimulate antitumor immune responses. OPTiM, a Phase III trial of T-VEC vs GM-CSF in unresectable stage IIIB-IV melanoma, improved the primary endpoint
- Published
- 2015
44. Directional motility induced by epidermal growth factor requires Cdc42
- Author
-
Akihiro Iwabu, Nancy A. Burke, Jeffrey Chou, Alan Wells, and Simon C. Watkins
- Subjects
Phosphatidylinositol 4,5-Diphosphate ,Cell ,Motility ,CDC42 ,Biology ,Gene Expression Regulation, Enzymologic ,Cell Line ,chemistry.chemical_compound ,Cell Movement ,Epidermal growth factor ,Cell polarity ,medicine ,Animals ,Pseudopodia ,Phosphatidylinositol ,cdc42 GTP-Binding Protein ,Epidermal Growth Factor ,Phospholipase C ,Phospholipase C gamma ,Cell Polarity ,Cell Biology ,Cell biology ,Eukaryotic Cells ,medicine.anatomical_structure ,chemistry ,Type C Phospholipases ,Intracellular - Abstract
Cell motility is actuated by a host of intracellular signaling cascades that result in movement of the cell in one direction, even without an external gradient. Phospholipase C-gamma (PLCgamma) has been shown to be important for growth factor-induced lamellipodial protrusion at the front of the cell while Cdc42 has been implicated in both filopodium formation at the leading edge and control of polarity of migrating cells. We asked whether these asymmetries in effector molecules may be linked. When we overexpressed either constitutively active, dominant negative, or GFP-tagged Cdc42, wild-type NR6 fibroblasts lost directionality, as expected. On epidermal growth factor (EGF) exposure these cells produced multiple, transient protrusions in every direction; these extensions failed to result in productive motility. GFP-tagged Cdc42 appeared transiently at edges of newly formed protrusions in EGF-stimulated cells while they moved haphazardly. While PLCgamma is distributed throughout the cell, the ratio of active, tyrosyl-phosphorylated PLCgamma was increased at the leading edge, where phosphatidylinositol (4,5)-bisphosphate (PIP(2)) hydrolysis is concentrated. This co-localization of activities may be due to Cdc42 directing PLCgamma to the cell front, as PLCgamma associated with Cdc42 in an EGF-dependent manner. We conclude that Cdc42 controls cell polarity, likely in part, through its binding to active PLCgamma.
- Published
- 2003
45. Safety, pharmacokinetics (PK) and pharmacodynamics (PD) data from a phase I dose-escalation study of OX40 agonistic monoclonal antibody (mAb) PF-04518600 (PF-8600) in combination with utomilumab, a 4-1BB agonistic mAb
- Author
-
Jeffrey S. Wasser, Ken Liao, Siwen Hu-Lieskovan, J-P. Spano, V. Dell, Eric Angevin, Tenshang Joh, Patrick A. Ott, Alberto Chiappori, Bishu J Ganguly, Toshihiko Doi, John A. Thompson, F. Eskens, Willeke Ros, Omid Hamid, Adi Diab, Naiyer A. Rizvi, Catherine Fleener, Anthony B. El-Khoueiry, and Jeffrey Chou
- Subjects
0301 basic medicine ,business.industry ,medicine.drug_class ,Hematology ,Pharmacology ,Monoclonal antibody ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Pharmacokinetics ,030220 oncology & carcinogenesis ,Pharmacodynamics ,Agonistic behaviour ,Dose escalation ,Medicine ,Utomilumab ,business - Published
- 2017
46. The relationship of pharmacodynamics (PD) and pharmacokinetics (PK) to clinical outcomes in a phase I study of OX40 agonistic monoclonal antibody (mAb) PF-04518600 (PF-8600)
- Author
-
Omid Hamid, John A. Thompson, Toshihiko Doi, Siwen Hu-Lieskovan, Ferry A.L.M. Eskens, Jeffrey Chou, Tenshang Joh, Ken Liao, Bishu J Ganguly, Anthony B. El-Khoueiry, Catherine Fleener, Willeke Ros, and Adi Diab
- Subjects
0301 basic medicine ,Cancer Research ,medicine.drug_class ,business.industry ,SUPERFAMILY ,Pharmacology ,Monoclonal antibody ,Phase i study ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Pharmacokinetics ,030220 oncology & carcinogenesis ,Pharmacodynamics ,medicine ,Agonistic behaviour ,business ,Tumor necrosis factor receptor - Abstract
3027 Background: PF-8600 is a novel fully human IgG2 agonistic mAb against human OX40, a TNF receptor superfamily member expressed primarily on activated T cells. This ongoing phase 1 study (NCT02315066) is investigating the safety, efficacy, PK and PD of PF-8600 in patients (pts) with solid tumors. Methods: PF-8600 (0.01–10 mg/kg) was given IV every 14d. Expression of free/total OX40 receptor, proliferation marker ki67 and activation markers HLA-DR/CD38 were measured in T cell subsets in peripheral blood by flow cytometry in all pts. CD4, CD8, OX40 and FOXP3 were evaluated in paired tumor biopsies (bx), collected from a subset of pts (≥0.1 mg/kg) at baseline (BL) and Wk6, by immunohistochemistry. Results: As of 21Sep2016, 48 pts with melanoma (n = 14), hepatocellular carcinoma (HCC, n = 19), head and neck squamous cell (n = 6) or renal cell carcinoma (n = 9) enrolled in the dose-escalation cohorts (0.01-3 mg/kg). No immune related adverse events (AE) were reported. The most frequent treatment related AEs in > 3 pts were fatigue (27.1%) and nausea and vomiting (8.3% each); all Gr 1-2. 2 pts had a partial response: melanoma at 0.1 (Pt1) and HCC at 0.3 (Pt2) mg/kg. 25 pts had best ORR (BOR) of stable disease (SD; 3 pts ≥24 wks). A majority of pts at 0.1, 0.3, and 3 mg/kg, including Pt1 and Pt2, had increases in ki67 and HLA-DR/CD38 expression in peripheral CD4+ central memory T cells. Pt1, Pt2 and all pts at ≥0.3 mg/kg had full receptor occupancy. Paired bx were only available from pts with BOR of SD or progressive disease. In 10 pts with available paired tumor bx and defined date of radiographic progression (rPD), longer time to rPD correlated with increases in %OX40+ in bx from BL to Wk6, regardless of dose level, tumor type or prior immunotherapy (R2= 0.52, p = 0.0188); no correlation between rPD and CD4+, CD8+ or FOXP3+ expression changes was observed. Updated efficacy, safety, PK and PD data will be presented. Conclusions: PF-8600 is well tolerated with evidence of single agent efficacy. Initial observations of PD markers that change with treatment and correlate with rPD support efforts to confirm these findings as more clinical outcomes and larger sample sizes become available. Clinical trial information: NCT02315066.
- Published
- 2017
47. Immune escape from NY-ESO-1-specific T-cell therapy via loss of heterozygosity in the MHC
- Author
-
Paul D. Robbins, Zandra K. Klippel, Lilien N. Voong, Edus H. Warren, Andrea M. H. Towlerton, Jeffrey Chou, and William I. Bensinger
- Subjects
Adoptive cell transfer ,Heterozygote ,medicine.medical_treatment ,T cell ,Antigen-Presenting Cells ,Mice, SCID ,Biology ,CD8-Positive T-Lymphocytes ,Major histocompatibility complex ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Antigen ,Mice, Inbred NOD ,Cell Line, Tumor ,HLA-A2 Antigen ,Genetics ,medicine ,Animals ,Humans ,NY-ESO-1 ,Antigen-presenting cell ,Molecular Biology ,Alleles ,immune escape ,Immunotherapy ,Peptide Fragments ,3. Good health ,Neoplasm Proteins ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,Cancer research ,Molecular Medicine ,Tumor Escape ,Multiple Myeloma ,adoptive immunotherapy ,CD8 ,030215 immunology - Abstract
Adoptive immunotherapy of tumors with T cells specific for the cancer-testis antigen NY-ESO-1 has shown great promise in preclinical models and in early stage clinical trials. Tumor persistence or recurrence after NY-ESO-1-specific therapy occurs, however, and the mechanisms of recurrence remain poorly defined. In a murine xenograft model of NY-ESO-1+ multiple myeloma, we observed tumor recurrence after adoptive transfer of CD8+ T cells genetically redirected to the prototypic NY-ESO-1157-165 peptide presented by HLA-A*02:01. Analysis of the myeloma cells that had escaped from T cell control revealed intact expression of NY-ESO-1 and B2M, but selective, complete loss of HLA-A*02:01 expression from the cell surface. Loss of heterozygosity in the Major Histocompatibility Complex (MHC) involving the HLA-A locus was identified in the tumor cells, and further analysis revealed selective loss of the allele encoding HLA-A*02:01. Although loss of heterozygosity involving the MHC has not been described in myeloma patients with persistent or recurrent disease after immune therapies such as allogeneic hematopoietic cell transplantation (HCT), it has been described in patients with acute myelogenous leukemia who relapsed after allogeneic HCT. These results suggest that MHC loss should be evaluated in patients with myeloma and other cancers who relapse after adoptive NY-ESO-1-specific T cell therapy.
- Published
- 2013
48. Phenotypic and transcriptional fidelity of patient-derived colon cancer xenografts in immune-deficient mice
- Author
-
Christie-Lynn Mortales, Matthew P. Fitzgibbon, Melissa P. Upton, Raymond S. Yeung, Martin W. McIntosh, Andrea M. H. Towlerton, Edus H. Warren, and Jeffrey Chou
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Stromal cell ,Colorectal cancer ,medicine.medical_treatment ,lcsh:Medicine ,Mice, SCID ,Biology ,Metastasis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cancer immunotherapy ,medicine ,Animals ,Humans ,lcsh:Science ,030304 developmental biology ,0303 health sciences ,Mixed tumor ,Multidisciplinary ,lcsh:R ,Immunotherapy ,medicine.disease ,3. Good health ,Haematopoiesis ,030220 oncology & carcinogenesis ,Colonic Neoplasms ,Female ,lcsh:Q ,Research Article - Abstract
Xenografts of human colorectal cancer (CRC) in immune-deficient mice have great potential for accelerating the study of tumor biology and therapy. We evaluated xenografts established in NOD/scid/IL2Rγ-null mice from the primary or metastatic tumors of 27 patients with CRC to estimate their capacity for expanding tumor cells for in vitro studies and to assess how faithfully they recapitulated the transcriptional profile of their parental tumors. RNA-seq analysis of parental human CRC tumors and their derivative xenografts demonstrated that reproducible transcriptional changes characterize the human tumor to murine xenograft transition. In most but not all cases, the human stroma, vasculature, and hematopoietic elements were systematically replaced by murine analogues while the carcinoma component persisted. Once established as xenografts, human CRC cells that could be propagated by serial transplantation remained transcriptionally stable. Three histologically atypical xenografts, established from patients with peritoneal metastases, contained abundant human stromal elements and blood vessels in addition to human tumor cells. The transcriptomes of these mixed tumor/stromal xenografts did not closely resemble those of their parental tumors, and attempts to propagate such xenografts by serial transplantation were unsuccessful. Stable expression of numerous genes previously identified as high priority targets for immunotherapy was observed in most xenograft lineages. Aberrant expression in CRC cells of human genes that are normally only expressed in hematopoietic cells was also observed. Our results suggest that human CRC cells expanded in murine xenografts have great utility for studies of tumor immunobiology and targeted therapies such as immunotherapy but also identify potential limitations.
- Published
- 2013
49. A phase 1/3 multicenter trial of talimogene laherparepvec in combination with pembrolizumab for unresected, stage IIIB-IV melanoma (MASTERKEY-265)
- Author
-
Scott J. Diede, Ari M. Vanderwalde, Antoni Ribas, Jeffrey Chou, F. Stephen Hodi, Reinhard Dummer, Anthony J. Olszanski, Thomas F. Gajewski, John M. Kirkwood, Jonathan Cebon, Olivier Michielin, Josep Malvehy, Igor Puzanov, Eugenio Fernandez, Olga M. Kuznetsova, Robert H.I. Andtbacka, Georgina V. Long, and Lisa Chen
- Subjects
Cancer Research ,business.industry ,medicine.medical_treatment ,Melanoma ,Immunotherapy ,Pembrolizumab ,medicine.disease ,Oncolytic virus ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,Unresected ,Lytic cycle ,030220 oncology & carcinogenesis ,Multicenter trial ,Cancer research ,medicine ,Talimogene laherparepvec ,business ,030215 immunology - Abstract
TPS9598Background: Talimogene laherparepvec, an oncolytic viral immunotherapy, was designed to selectively replicate in tumors resulting in lytic cell death, antigen release, and production of GM-C...
- Published
- 2016
50. Efficacy analysis of MASTERKEY-265 phase 1b study of talimogene laherparepvec (T-VEC) and pembrolizumab (pembro) for unresectable stage IIIB-IV melanoma
- Author
-
Abraham Anderson, David Ross Kaufman, Christine K. Gause, John M. Kirkwood, Robert H.I. Andtbacka, Jonathan Cebon, Thomas F. Gajewski, Antoni Ribas, Jeffrey Chou, F. Stephen Hodi, Anthony J. Olszanski, Lisa Chen, Kevin S. Gorski, Ari M. Vanderwalde, Georgina V. Long, Reinhard Dummer, Olivier Michielin, Igor Puzanov, Eugenio Fernandez, and Josep Malvehy
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,business.industry ,medicine.medical_treatment ,Melanoma ,Immunotherapy ,Pembrolizumab ,medicine.disease_cause ,medicine.disease ,Oncolytic virus ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Herpes simplex virus ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Biomarker (medicine) ,Stage (cooking) ,Talimogene laherparepvec ,business - Abstract
9568Background: T-VEC is a herpes simplex virus (HSV)-1 -based oncolytic immunotherapy designed to selectively replicate in tumors, produce GM-CSF and stimulate antitumor immune responses in melanoma. T-VEC significantly improved durable response rate vs GM-CSF in stage IIIB-IV melanoma patients (pts) with injectable tumors. Pembro inhibits programmed cell death protein 1 and improves survival in advanced melanoma. The combination may further improve clinical benefit. Here we report phase 1b efficacy, safety and biomarker data from a phase 1b/3 study of T-VEC+pembro in unresectable stage IIIB-IV melanoma (NCT02263508) with all pts having started on T-VEC+pembro ≥ 6 mo prior. Methods: Key inclusion criteria: unresectable stage IIIB-IV melanoma, injectable lesions; no prior systemic tx; and ECOG PS 0-1. T-VEC: ≤ 4 mL in (sub)cutaneous/nodal lesions, 106 PFU/mL d1, 108PFU/mL d22 then Q2W; pembro: IV, 200 mg d36 then Q2W. Tx until first occurrence of: complete response (CR); no injectable tumors (for T-VEC); ...
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.