5901
|
Hamanishi J, Mandai M, Konishi I. Immune checkpoint inhibition in ovarian cancer. Int Immunol 2016; 28:339-48. [PMID: 27055470 DOI: 10.1093/intimm/dxw020] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 04/05/2016] [Indexed: 12/13/2022] Open
Abstract
Recent studies have shown that tumor cells acquire escape mechanisms to evade host immunity in the tumor microenvironment. Two key immune checkpoint pathways mediated by immunosuppressive co-signaling, the first via programmed cell death 1 (PD-1) and PD-1 ligand 1 (PD-1/PD-L1) and the second via CTLA-4 and B7 (CTLA-4/B7), have been previously described. Several clinical trials have revealed an outstanding anti-tumor efficacy of immune checkpoint inhibitors (anti-CTLA-4 antibody, anti-PD-1 antibody and/or anti-PD-L1 antibody) in patients with various types of solid malignancies, including non-small cell lung cancer, melanoma, renal cell cancer and ovarian cancer. In this review, we examine pre-clinical studies that described the local immune status and immune checkpoint signals in ovarian cancer, highlight recent clinical trials that evaluated immune checkpoint inhibitors against ovarian cancer and discuss the clinical issues regarding immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, 54 Shogoinkawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Masaki Mandai
- Department of Obstetrics and Gynecology, Kinki University, Higashiosaka, Osaka, 577-8502, Japan
| | - Ikuo Konishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, 54 Shogoinkawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
5902
|
Volejnikova J, Bajciova V, Sulovska L, Geierova M, Buriankova E, Jarosova M, Hajduch M, Sterba J, Mihal V. Bone marrow metastasis of malignant melanoma in childhood arising within a congenital melanocytic nevus. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2016; 160:456-60. [PMID: 27049531 DOI: 10.5507/bp.2016.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 03/23/2016] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Malignant melanoma in childhood is infrequent and can arise within congenital melanocytic nevi. Spread of malignant melanoma to the bone marrow, especially in children, is extremely rare. METHODS AND RESULTS Reported is a case of a 5-year-old boy with a congenital large melanocytic nevus of the head and neck who presented with a short history of low back and leg pain, fever and cervical lymphadenopathy. Despite regular follow-up by a dermatologist and plastic surgeon and repeatedly negative histology of previous partial excisions, diffuse bone marrow infiltration with malignant melanoma was diagnosed. The primary site was identified in the post-excision area. The disease progressed rapidly on ipilimumab immunotherapy and led to death at four months from the diagnosis. CONCLUSION Surveillance is indispensable in children with a predisposition to melanoma and nonspecific symptoms such as bone pain, gait impairment or cytopenia, should always be taken into account.
Collapse
Affiliation(s)
- Jana Volejnikova
- Department of Pediatrics, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Czech Republic.,Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic
| | - Viera Bajciova
- Department of Pediatric Oncology, Masaryk University, Brno and University Hospital Brno, Czech Republic
| | - Lucie Sulovska
- Department of Pediatrics, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Czech Republic
| | - Marie Geierova
- Department of Clinical and Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Czech Republic
| | - Eva Buriankova
- Department of Nuclear Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Czech Republic
| | - Marie Jarosova
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Czech Republic
| | - Marian Hajduch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic
| | - Jaroslav Sterba
- Department of Pediatric Oncology, Masaryk University, Brno and University Hospital Brno, Czech Republic
| | - Vladimir Mihal
- Department of Pediatrics, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Czech Republic.,Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic
| |
Collapse
|
5903
|
Buonerba C, Di Lorenzo G, Sonpavde G. Combination therapy for metastatic renal cell carcinoma. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:100. [PMID: 27047959 DOI: 10.21037/atm.2016.01.31] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Current therapy for metastatic clear cell renal cell carcinoma (RCC) consists of the serial administration of single agents. Combinations of VEGF and mTOR inhibitors have been disappointing in previous randomized trials. However, the combination of lenvatinib, a multitargeted agent that inhibits VEGF as well as FGF receptors, and everolimus demonstrated promising results in a randomized phase II trial. Moreover, the emergence of programmed cell death 1 (PD-1) and programmed cell death ligand 1 (PD-L1) inhibitors has spawned the investigation of combinations of these agents with VEGF inhibitors and cytotoxic T-lymphocyte antigen 4 (CTLA-4) inhibitors. These ongoing phase III trials in conjunction with the development of predictive biomarkers and agents inhibiting novel therapeutic targets may provide much needed advances in this still largely incurable disease.
Collapse
Affiliation(s)
- Carlo Buonerba
- 1 University Federico II, Naples, Italy, 2 Comprehensive Cancer Center, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Giuseppe Di Lorenzo
- 1 University Federico II, Naples, Italy, 2 Comprehensive Cancer Center, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Guru Sonpavde
- 1 University Federico II, Naples, Italy, 2 Comprehensive Cancer Center, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| |
Collapse
|
5904
|
Chae YK, Pan A, Davis AA, Raparia K, Mohindra NA, Matsangou M, Giles FJ. Biomarkers for PD-1/PD-L1 Blockade Therapy in Non-Small-cell Lung Cancer: Is PD-L1 Expression a Good Marker for Patient Selection? Clin Lung Cancer 2016; 17:350-361. [PMID: 27137346 DOI: 10.1016/j.cllc.2016.03.011] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/23/2016] [Accepted: 03/29/2016] [Indexed: 12/14/2022]
Abstract
Immunotherapy has emerged as a promising treatment modality in cancer therapy. With improved understanding of how to tip the balance of immune homeostasis, novel therapeutics targeting immune checkpoints have been developed, with durable responses observed in multiple solid tumors, including melanoma, renal cell carcinoma, and non-small-cell lung cancer. Clinical trials have reported favorable responses using programmed cell death-1 protein receptor (PD-1)/programmed cell death-1 protein ligand (PD-L1) blockade as monotherapy and most impressively in combinatorial trials with cytotoxic T-lymphocyte antigen-4 protein blockade. Nonetheless, a clinical benefit has not been observed in all patients. Therefore, identifying the ideal biomarkers for patient selection would be of great value in optimizing and personalizing immunotherapy. The utility of PD-L1 expression as a biomarker has varied in different clinical trials and immunohistochemistry assays. In addition, the response to immune checkpoint inhibition has been complicated by PD-L1 expression as a marker influenced by the dynamic tumor microenvironment. No consensus has yet been reached on whether PD-L1 expression is an ideal marker for patient selection. Recent research has shown promise for alternative markers, including T-cell immunohistochemistry, other immunologic markers, T-cell receptor clonality, and somatic mutational burden. However, additional studies are needed to assess the value of these as practical predictive biomarkers for patient selection and treatment response.
Collapse
Affiliation(s)
- Young Kwang Chae
- Developmental Therapeutics Program of the Division of Hematology Oncology, Department of Medicine, Northwestern Medicine Developmental Therapeutics Institute, Northwestern University, Chicago, IL; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL; Northwestern University Feinberg School of Medicine, Chicago, IL.
| | - Alan Pan
- Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Andrew A Davis
- Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Kirtee Raparia
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL; Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Nisha A Mohindra
- Developmental Therapeutics Program of the Division of Hematology Oncology, Department of Medicine, Northwestern Medicine Developmental Therapeutics Institute, Northwestern University, Chicago, IL; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL; Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Maria Matsangou
- Developmental Therapeutics Program of the Division of Hematology Oncology, Department of Medicine, Northwestern Medicine Developmental Therapeutics Institute, Northwestern University, Chicago, IL; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL; Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Francis J Giles
- Developmental Therapeutics Program of the Division of Hematology Oncology, Department of Medicine, Northwestern Medicine Developmental Therapeutics Institute, Northwestern University, Chicago, IL; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL; Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
5905
|
Riaz N, Morris L, Havel JJ, Makarov V, Desrichard A, Chan TA. The role of neoantigens in response to immune checkpoint blockade. Int Immunol 2016; 28:411-9. [PMID: 27048318 DOI: 10.1093/intimm/dxw019] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 03/30/2016] [Indexed: 12/17/2022] Open
Abstract
Immune checkpoint blockade has demonstrated substantial promise for the treatment of several advanced malignancies. These agents activate the immune system to attack tumor cells. For example, agents targeting CTLA4 and programmed cell death 1 (PD-1) have resulted in impressive response rates and, in some cases, durable remissions. Neoantigens are mutations that encode immunologically active proteins that can cause the immune system to recognize the affected cell as foreign. Recent data have made it clear that these mutations are, in large part, the functional targets of immune checkpoint blockade. This review summarizes the key discoveries leading up to this important conclusion and discusses possible applications of neoantigens in cancer therapy.
Collapse
Affiliation(s)
| | - Luc Morris
- Human Oncology and Pathogenesis Program and Department of Surgery, Memorial Sloan Kettering Cancer Center, Box 20, 1275 York Avenue, New York, NY 10065, USA
| | | | | | | | - Timothy A Chan
- Department of Radiation Oncology, Human Oncology and Pathogenesis Program and
| |
Collapse
|
5906
|
Graham L, Schweizer MT. Targeting persistent androgen receptor signaling in castration-resistant prostate cancer. Med Oncol 2016; 33:44. [DOI: 10.1007/s12032-016-0759-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 03/24/2016] [Indexed: 12/19/2022]
|
5907
|
Gros A, Parkhurst MR, Tran E, Pasetto A, Robbins PF, Ilyas S, Prickett TD, Gartner JJ, Crystal JS, Roberts IM, Trebska-McGowan K, Wunderlich JR, Yang JC, Rosenberg SA. Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat Med 2016; 22:433-8. [PMID: 26901407 PMCID: PMC7446107 DOI: 10.1038/nm.4051] [Citation(s) in RCA: 664] [Impact Index Per Article: 73.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/26/2016] [Indexed: 01/10/2023]
Abstract
Detection of lymphocytes that target tumor-specific mutant neoantigens--derived from products encoded by mutated genes in the tumor--is mostly limited to tumor-resident lymphocytes, but whether these lymphocytes often occur in the circulation is unclear. We recently reported that intratumoral expression of the programmed cell death 1 (PD-1) receptor can guide the identification of the patient-specific repertoire of tumor-reactive CD8(+) lymphocytes that reside in the tumor. In view of these findings, we investigated whether PD-1 expression on peripheral blood lymphocytes could be used as a biomarker to detect T cells that target neoantigens. By using a high-throughput personalized screening approach, we identified neoantigen-specific lymphocytes in the peripheral blood of three of four melanoma patients. Despite their low frequency in the circulation, we found that CD8(+)PD-1(+), but not CD8(+)PD-1(-), cell populations had lymphocytes that targeted 3, 3 and 1 unique, patient-specific neoantigens, respectively. We show that neoantigen-specific T cells and gene-engineered lymphocytes expressing neoantigen-specific T cell receptors (TCRs) isolated from peripheral blood recognized autologous tumors. Notably, the tumor-antigen specificities and TCR repertoires of the circulating and tumor-infiltrating CD8(+)PD-1(+) cells appeared similar, implying that the circulating CD8(+)PD-1(+) lymphocytes could provide a window into the tumor-resident antitumor lymphocytes. Thus, expression of PD-1 identifies a diverse and patient-specific antitumor T cell response in peripheral blood, providing a novel noninvasive strategy to develop personalized therapies using neoantigen-reactive lymphocytes or TCRs to treat cancer.
Collapse
MESH Headings
- Adult
- Aged
- Antigens, Neoplasm/blood
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cell Line, Tumor
- Female
- Humans
- Immunotherapy
- Lymphocytes/immunology
- Lymphocytes/pathology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/pathology
- Male
- Melanoma/blood
- Melanoma/genetics
- Melanoma/immunology
- Melanoma/therapy
- Middle Aged
- Programmed Cell Death 1 Receptor/blood
- Programmed Cell Death 1 Receptor/genetics
- Programmed Cell Death 1 Receptor/immunology
- Prospective Studies
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
Collapse
Affiliation(s)
- Alena Gros
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Maria R Parkhurst
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Eric Tran
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Anna Pasetto
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Paul F Robbins
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Sadia Ilyas
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Todd D Prickett
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Jared J Gartner
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Jessica S Crystal
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Ilana M Roberts
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Kasia Trebska-McGowan
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - John R Wunderlich
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - James C Yang
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| | - Steven A Rosenberg
- Surgery Branch, National Cancer Institute (NCI), National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
5908
|
Successful Immunotherapy against a Transplantable Mouse Squamous Lung Carcinoma with Anti–PD-1 and Anti-CD137 Monoclonal Antibodies. J Thorac Oncol 2016; 11:524-36. [DOI: 10.1016/j.jtho.2016.01.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 12/07/2015] [Accepted: 01/01/2016] [Indexed: 11/23/2022]
|
5909
|
Abstract
Recent data have demonstrated improved survival with targeted and immune therapies in patients with advanced melanoma, leading to much excitement amongst the oncology community and the widespread use of these drugs in combination regimens. However, the place of these combination therapies in the treatment of advanced melanoma remains to be fully determined. In this perspectives article, we critically review the available data and outline the rationale for these combinations being adopted as the standard of care for patients with advanced melanoma in the future.
Collapse
Affiliation(s)
- Keiran S M Smalley
- The Department of Cutaneous Oncology, Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, USA.
- The Department of Tumor Biology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| | - Zeynep Eroglu
- The Department of Cutaneous Oncology, Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Vernon K Sondak
- The Department of Cutaneous Oncology, Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| |
Collapse
|
5910
|
Schiavone MB, Broach V, Shoushtari AN, Carvajal RD, Alektiar K, Kollmeier MA, Abu-Rustum NR, Leitao MM. Combined immunotherapy and radiation for treatment of mucosal melanomas of the lower genital tract. Gynecol Oncol Rep 2016; 16:42-6. [PMID: 27331137 PMCID: PMC4899413 DOI: 10.1016/j.gore.2016.04.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 04/06/2016] [Accepted: 04/10/2016] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVE To report our experience using ipilimumab, a monoclonal antibody targeting CTLA-4, combined with radiation therapy in women diagnosed with mucosal melanoma of the lower genital tract. METHODS We retrospectively identified all patients who received ipilimumab with concurrent radiation treatment of mucosal melanoma of the lower genital tract at Memorial Sloan Kettering Cancer Center from 2012 to 2015. Various clinicopathologic data and treatment response were abstracted and analyzed. RESULTS Four patients were identified. Median age was 61.5 years (range 44-68); 3 were diagnosed with vaginal melanoma, 1 with cervical melanoma. All would have required extensive surgical procedures to remove entirety of disease. Median size of lesions was 4.7 cm (range, 3.3-5.3); all were Ballantyne stage I. Median number of doses of upfront ipilimumab was 4 (range, 3-4). Two patients suffered CTCAE grade 3 adverse events (colitis, rash). All received external beam radiation: 3 to 3000 cGy, 1 to 6020 cGy. Post-radiation surgical resection was performed in 3 patients (75%); 1 (33%) of 3 patients achieved complete pathologic response. Complete local radiographic response was observed in all patients after completion of initial therapy and surgery. Two developed recurrence at 9 and 10 months post-diagnosis (mediastinum, lung); 2 remain disease-free at 20 and 38 months. CONCLUSIONS Mucosal melanoma of the lower genital tract is rare, and data-driven treatment strategies limited. Immunotherapy has demonstrated durable efficacy in the treatment of cutaneous melanomas. Our small case series shows a favorable response to combined ipilimumab and radiation therapy. Larger studies are needed to validate these promising results.
Collapse
Affiliation(s)
- Maria B. Schiavone
- Gynecologic Oncology, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
| | - Vance Broach
- Gynecologic Oncology, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
| | - Alexander N. Shoushtari
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
- Weill Cornell Medical College, 1300 York Avenue, New York, NY, USA
| | - Richard D. Carvajal
- Experimental Therapeutics, Division of Hematology/Oncology, Columbia University Medical Center, 161 Fort Washington Avenue, New York, NY, USA
- Melanoma Service, Division of Hematology/Oncology, Columbia University Medical Center, 161 Fort Washington Avenue, New York, NY, USA
| | - Kaled Alektiar
- Weill Cornell Medical College, 1300 York Avenue, New York, NY, USA
- Radiation Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
| | - Marisa A. Kollmeier
- Weill Cornell Medical College, 1300 York Avenue, New York, NY, USA
- Radiation Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
| | - Nadeem R. Abu-Rustum
- Gynecologic Oncology, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
- Weill Cornell Medical College, 1300 York Avenue, New York, NY, USA
| | - Mario M. Leitao
- Gynecologic Oncology, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA
- Weill Cornell Medical College, 1300 York Avenue, New York, NY, USA
| |
Collapse
|
5911
|
Vormehr M, Diken M, Boegel S, Kreiter S, Türeci Ÿ, Sahin U. Mutanome directed cancer immunotherapy. Curr Opin Immunol 2016; 39:14-22. [DOI: 10.1016/j.coi.2015.12.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 12/01/2015] [Accepted: 12/02/2015] [Indexed: 10/22/2022]
|
5912
|
Kaliks RA. An update on clinical oncology for the non-oncologist. EINSTEIN-SAO PAULO 2016; 14:294-9. [PMID: 27462901 PMCID: PMC4943365 DOI: 10.1590/s1679-45082016md3550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 11/13/2015] [Indexed: 11/22/2022] Open
Abstract
ABSTRACTRecent advances in the understanding of tumor driver mutations, signaling pathways that lead to tumor progression, and the better understanding of the interaction between tumor cells and the immune system are revolutionizing cancer treatment. The pace at which new treatments are approved and the prices at which they are set have made it even more difficult to offer these treatments in countries like Brazil. In this review we present for the non-oncologist these new treatments and compare their availability in Brazilian public health system and private health system with that of developed countries.RESUMOAvanços recentes na compreensão de mutações promotoras de desenvolvimento do câncer, sinalização que leva à progressão de tumores, e o avanço no entendimento da interação entre as células tumorais e o sistema imunológico estão revolucionando o tratamento do câncer. A velocidade com que novos tratamentos são aprovados e o alto custo das medicações dificultam a disponibilização de terapêuticas em países como o Brasil. Nesta revisão, apresentamos ao não oncologista esses novos tratamentos e comparamos sua disponibilidade nos sistemas público e privado de saúde no Brasil com os países desenvolvidos.
Collapse
|
5913
|
Friedman CF, Postow MA. Emerging Tissue and Blood-Based Biomarkers that may Predict Response to Immune Checkpoint Inhibition. Curr Oncol Rep 2016; 18:21. [PMID: 26922327 PMCID: PMC5063308 DOI: 10.1007/s11912-016-0509-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The immune system plays an essential role in the surveillance and eradication of neoplastic cells. This interaction is modulated via immunologic regulators (checkpoints). Antibodies that block the checkpoints cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), and the programmed cell death protein 1 pathway (PD1/PD-L1) have demonstrated efficacy in a number of malignancies. However, response rates are variable, and administration of these antibodies can be associated with immune-related adverse events. Therefore, researchers are engaged in an effort to discover biomarkers that may predict response to these agents. This review focuses on potential blood and tumor-based biomarkers that have been assessed in patients treated with these checkpoint-blocking antibodies.
Collapse
Affiliation(s)
- Claire F Friedman
- Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA.
| | - Michael A Postow
- Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA.
| |
Collapse
|
5914
|
Weber J, Gibney G, Kudchadkar R, Yu B, Cheng P, Martinez AJ, Kroeger J, Richards A, McCormick L, Moberg V, Cronin H, Zhao X, Schell M, Chen YA. Phase I/II Study of Metastatic Melanoma Patients Treated with Nivolumab Who Had Progressed after Ipilimumab. Cancer Immunol Res 2016; 4:345-53. [PMID: 26873574 PMCID: PMC4818672 DOI: 10.1158/2326-6066.cir-15-0193] [Citation(s) in RCA: 210] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 01/12/2016] [Indexed: 11/16/2022]
Abstract
The checkpoint inhibitor nivolumab is active in patients with metastatic melanoma who have failed ipilimumab. In this phase I/II study, we assessed nivolumab's safety in 92 ipilimumab-refractory patients with unresectable stage III or IV melanoma, including those who experienced grade 3-4 drug-related toxicity to ipilimumab. We report long-term survival, response duration, and biomarkers in these patients after nivolumab treatment (3 mg/kg) every 2 weeks for 24 weeks, then every 12 weeks for up to 2 years, with or without a multipeptide vaccine. The response rate for ipilimumab-refractory patients was 30% (95% CI, 21%-41%). The median duration of response was 14.6 months, median progression-free survival was 5.3 months, and median overall survival was 20.6 months, when patients were followed up for a median of 16 months. One- and 2-year survival rates were 68.4% and 31.2%, respectively. Ipilimumab-naïve and ipilimumab-refractory patients showed no significant difference in survival. The 21 patients with prior grade 3-4 toxicity to ipilimumab that was managed with steroids tolerated nivolumab well, with 62% (95% CI, 38%-82%) having complete or partial responses or stabilized disease at 24 weeks. High numbers of myeloid-derived suppressor cells (MDSC) were associated with poor survival. Thus, survival and long-term safety were excellent in ipilimumab-refractory patients treated with nivolumab. Prior grade 3-4 immune-related adverse effects from ipilimumab were not indicative of nivolumab toxicities, and patients had a high overall rate of remission or stability at 24 weeks. Prospectively evaluating MDSC numbers before treatment could help assess the expected benefit of nivolumab.
Collapse
Affiliation(s)
- Jeffrey Weber
- Comprehensive Melanoma Research Center and Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida.
| | - Geoffrey Gibney
- Comprehensive Melanoma Research Center and Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Ragini Kudchadkar
- Comprehensive Melanoma Research Center and Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Bin Yu
- Comprehensive Melanoma Research Center and Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Pingyan Cheng
- Comprehensive Melanoma Research Center and Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Alberto J Martinez
- Comprehensive Melanoma Research Center and Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Jodie Kroeger
- Comprehensive Melanoma Research Center and Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Allison Richards
- Comprehensive Melanoma Research Center and Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Lori McCormick
- Comprehensive Melanoma Research Center and Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Valerie Moberg
- Comprehensive Melanoma Research Center and Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Heather Cronin
- Comprehensive Melanoma Research Center and Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Xiuhua Zhao
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida
| | - Michael Schell
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida
| | - Yian Ann Chen
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida
| |
Collapse
|
5915
|
Morrissey KM, Yuraszeck TM, Li C, Zhang Y, Kasichayanula S. Immunotherapy and Novel Combinations in Oncology: Current Landscape, Challenges, and Opportunities. Clin Transl Sci 2016; 9:89-104. [PMID: 26924066 PMCID: PMC5351311 DOI: 10.1111/cts.12391] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 02/22/2016] [Accepted: 02/22/2016] [Indexed: 12/11/2022] Open
Affiliation(s)
- KM Morrissey
- Department of Clinical PharmacologyGenentech IncSouth San FranciscoCaliforniaUSA
| | - TM Yuraszeck
- Clinical PharmacologyModeling and Simulation, Amgen IncThousand OaksCaliforniaUSA
| | - C‐C Li
- Department of Clinical PharmacologyGenentech IncSouth San FranciscoCaliforniaUSA
| | - Y Zhang
- Clinical PharmacologyModeling and Simulation, Amgen IncThousand OaksCaliforniaUSA
| | - S Kasichayanula
- Clinical PharmacologyModeling and Simulation, Amgen IncThousand OaksCaliforniaUSA
| |
Collapse
|
5916
|
Zamarin D, Jazaeri AA. Leveraging immunotherapy for the treatment of gynecologic cancers in the era of precision medicine. Gynecol Oncol 2016; 141:86-94. [PMID: 27016233 PMCID: PMC5007873 DOI: 10.1016/j.ygyno.2015.12.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 11/25/2015] [Accepted: 12/30/2015] [Indexed: 12/14/2022]
Abstract
During the past decade significant progress in the understanding of stimulatory and inhibitory signaling pathways in immune cells has reinvigorated the field of immuno-oncology. In this review we outline the current immunotherapy based approaches for the treatment of gynecological cancers, and focus on the emerging clinical data on immune checkpoint inhibitors, adoptive cell therapies, and vaccines. It is anticipated that in the coming years biomarker-guided clinical trials, will provide for a better understanding of the mechanisms of response and resistance to immunotherapy, and guide combination treatment strategies that will extend the benefit from immunotherapy to patients with gynecologic cancers.
Collapse
Affiliation(s)
- Dmitriy Zamarin
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, United States
| | - Amir A Jazaeri
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas, MD Anderson Cancer Center, United States.
| |
Collapse
|
5917
|
The State of Cancer Care in America, 2016: A Report by the American Society of Clinical Oncology. J Oncol Pract 2016; 12:339-83. [PMID: 26979926 PMCID: PMC5015451 DOI: 10.1200/jop.2015.010462] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
5918
|
van der Burg SH, Arens R, Ossendorp F, van Hall T, Melief CJM. Vaccines for established cancer: overcoming the challenges posed by immune evasion. Nat Rev Cancer 2016; 16:219-33. [PMID: 26965076 DOI: 10.1038/nrc.2016.16] [Citation(s) in RCA: 524] [Impact Index Per Article: 58.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Therapeutic vaccines preferentially stimulate T cells against tumour-specific epitopes that are created by DNA mutations or oncogenic viruses. In the setting of premalignant disease, carcinoma in situ or minimal residual disease, therapeutic vaccination can be clinically successful as monotherapy; however, in established cancers, therapeutic vaccines will require co-treatments to overcome immune evasion and to become fully effective. In this Review, we discuss the progress that has been made in overcoming immune evasion controlled by tumour cell-intrinsic factors and the tumour microenvironment. We summarize how therapeutic benefit can be maximized in patients with established cancers by improving vaccine design and by using vaccines to increase the effects of standard chemotherapies, to establish and/or maintain tumour-specific T cells that are re-energized by checkpoint blockade and other therapies, and to sustain the antitumour response of adoptively transferred T cells.
Collapse
Affiliation(s)
| | - Ramon Arens
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | | | - Cornelis J M Melief
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
- ISA Pharmaceuticals, J. H. Oortweg 19, 2333 CH, Leiden, The Netherlands
| |
Collapse
|
5919
|
Bedognetti D, Maccalli C, Bader SBA, Marincola FM, Seliger B. Checkpoint Inhibitors and Their Application in Breast Cancer. Breast Care (Basel) 2016; 11:108-15. [PMID: 27239172 PMCID: PMC4881248 DOI: 10.1159/000445335] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Immune checkpoints are crucial for the maintenance of self-tolerance and for the modulation of immune responses in order to minimize tissue damage. Tumor cells take advantage of these mechanisms to evade immune recognition. A significant proportion of tumors, including breast cancers, can express co-inhibitory molecules that are important formediating the escape from T cell-mediated immune surveillance. The interaction of inhibitory receptors with their ligands can be blocked by specific molecules. Monoclonal antibodies (mAbs) directed against the cytotoxic T lymphocyte-associated antigen-4 (CTLA4) and, more recently, against the programmed cell death protein 1 (PD1), have been approved for the therapy of melanoma (anti-CTLA4 and anti-PD1 mAbs) and non-small cell lung cancer (anti-PD1 mAbs). Moreover, inhibition of PD1 signaling has shown extremely promising signs of activity in breast cancer. An increasing number of molecules directed against other immune checkpoints are currently under clinical development. In this review, we summarize the evidence supporting the implementation of checkpoint inhibition in breast cancer by reviewing in detail data on PD-L1 expression and its regulation. In addition, opportunities to boost anti-tumor immunity in breast cancer with checkpoint inhibitor-based immunotherapies alone and in combination with other treatment options will be discussed.
Collapse
Affiliation(s)
- Davide Bedognetti
- Tumor Biology, Immunology, and Therapy Section, Division of Translational Medicine, Sidra Medical and Research Center, Doha, Qatar
| | - Cristina Maccalli
- Tumor Biology, Immunology, and Therapy Section, Division of Translational Medicine, Sidra Medical and Research Center, Doha, Qatar
| | - Salha B.J. Al Bader
- National Center for Cancer Care and Research (NCCCR), and Hamad General Hospital, Doha, Qatar
| | - Francesco M. Marincola
- Office of the Chief Research Officer (CRO), Sidra Medical and Research Center, Doha, Qatar
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle/Saale, Germany
| |
Collapse
|
5920
|
|
5921
|
Abstract
Tumour metastasis, the movement of tumour cells from a primary site to progressively colonize distant organs, is a major contributor to the deaths of cancer patients. Therapeutic goals are the prevention of an initial metastasis in high-risk patients, shrinkage of established lesions and prevention of additional metastases in patients with limited disease. Instead of being autonomous, tumour cells engage in bidirectional interactions with metastatic microenvironments to alter antitumour immunity, the extracellular milieu, genomic stability, survival signalling, chemotherapeutic resistance and proliferative cycles. Can targeting of these interactions significantly improve patient outcomes? In this Review preclinical research, combination therapies and clinical trial designs are re-examined.
Collapse
Affiliation(s)
- Patricia S Steeg
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| |
Collapse
|
5922
|
Affiliation(s)
- Anthony J Olszanski
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, and The Angeles Clinic and Research Institute, Los Angeles, California
| | - Brianna W Hoffner
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, and The Angeles Clinic and Research Institute, Los Angeles, California
| |
Collapse
|
5923
|
Learning from the "tsunami" of immune checkpoint inhibitors in 2015. Crit Rev Oncol Hematol 2016; 101:213-20. [PMID: 27051042 DOI: 10.1016/j.critrevonc.2016.03.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/15/2016] [Accepted: 03/22/2016] [Indexed: 01/31/2023] Open
Abstract
2015 was marked by the tsunami of immune checkpoint inhibitors revealed by numerous FDA approvals, publications and abstracts in relation with these drugs in different cancers and settings. First, we reported all new indications of anti-CTLA4 and anti-PD1 approved by the FDA, the positive clinical trials published and the abstracts with promising results at important scientific meetings during 2015. Then, we discussed different critical issues of these new agents going from their predictive factors, combination therapies, tumor response patterns, efficacy in particular settings, side effect management to cost and economic burden.
Collapse
|
5924
|
Shu CA, Rizvi NA. Into the Clinic With Nivolumab and Pembrolizumab. Oncologist 2016; 21:527-8. [PMID: 27026678 PMCID: PMC4861376 DOI: 10.1634/theoncologist.2016-0099] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 03/14/2016] [Indexed: 12/26/2022] Open
Abstract
In 2015, the US Food and Drug Administration approved the use of nivolumab for treatment of squamous and nonsquamous non-small cell lung cancer (NSCLC) and pembrolizumab for use in patients with NSCLC tumors that express PD-L1. This commentary discusses issues with these two drugs relative to best practices and target populations.
Collapse
Affiliation(s)
- Catherine A Shu
- Division of Hematology/Oncology, Columbia University Medical Center, New York, New York, USA
| | - Naiyer A Rizvi
- Division of Hematology/Oncology, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
5925
|
Chen KH, Yuan CT, Tseng LH, Shun CT, Yeh KH. Case report: mismatch repair proficiency and microsatellite stability in gastric cancer may not predict programmed death-1 blockade resistance. J Hematol Oncol 2016; 9:29. [PMID: 27012666 PMCID: PMC4806434 DOI: 10.1186/s13045-016-0259-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 03/15/2016] [Indexed: 12/19/2022] Open
Abstract
Background Anti-programmed death-1 therapy has poor efficacy in mismatch repair-proficient (pMMR) colorectal cancers; however, its efficacy in pMMR gastric cancers remains undetermined. Here, we report the case of a patient with pMMR and microsatellite-stable gastric cancer who exhibited a partial response to salvage anti-programmed death-1 therapy with pembrolizumab. Case presentation Initially, the patient underwent subtotal gastrectomy 4 years ago for early-stage gastric cancer (pT1bN2M0, stage IIA). Immunohistochemical analysis of the tumor revealed strongly positive for HER2/neu. He had received trastuzumab plus pertuzumab, cisplatin, and capecitabine for recurrent tumors since September 2014 for 15 cycles. Disease progression of gastric cancer was found in August 2015. Since September 2015, the patient has received pembrolizumab monotherapy (200 mg as a fixed dose, every 3 weeks) for 3 months and the repeat computed tomography demonstrated a confirmed partial response. The plasma carcinoembryonic antigen also decreased dramatically. Both immunohistochemistry and a polymerase chain reaction-based method revealed that the patient had pMMR gastric cancer. Conclusions This case report provides the first report that mismatch repair-proficient and microsatellite-stable gastric cancers can respond well to anti-PD-1 monotherapy and indicates both markers may not sufficiently be predictive of anti-PD-1 therapy resistance in gastric cancer.
Collapse
Affiliation(s)
- Kuo-Hsing Chen
- Department of Oncology, National Taiwan University Hospital, 7, Chun-Shan S Rd, Taipei, 10002, Taiwan.,National Taiwan University Cancer Center, Taipei, Taiwan.,Graduate Institute of Oncology, National Taiwan University, Taipei, Taiwan
| | - Chang-Tsu Yuan
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Li-Hui Tseng
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Chia-Tung Shun
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan.,Department of Forensic Medicine, National Taiwan University, Taipei, Taiwan.,Forensic Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kun-Huei Yeh
- Department of Oncology, National Taiwan University Hospital, 7, Chun-Shan S Rd, Taipei, 10002, Taiwan. .,Graduate Institute of Oncology, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
5926
|
McLachlan J, Lima JPDSN, Dumas L, Banerjee S. Targeted agents and combinations in ovarian cancer: where are we now? Expert Rev Anticancer Ther 2016; 16:441-54. [DOI: 10.1586/14737140.2016.1162101] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
5927
|
Spagnolo F, Picasso V, Spano L, Tanda E, Venzano C, Queirolo P. Update on Metastatic Uveal Melanoma: Progress and Challenges. BioDrugs 2016; 30:161-72. [DOI: 10.1007/s40259-016-0167-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
5928
|
Dizon DS, Krilov L, Cohen E, Gangadhar T, Ganz PA, Hensing TA, Hunger S, Krishnamurthi SS, Lassman AB, Markham MJ, Mayer E, Neuss M, Pal SK, Richardson LC, Schilsky R, Schwartz GK, Spriggs DR, Villalona-Calero MA, Villani G, Masters G. Clinical Cancer Advances 2016: Annual Report on Progress Against Cancer From the American Society of Clinical Oncology. J Clin Oncol 2016; 34:987-1011. [PMID: 26846975 PMCID: PMC5075244 DOI: 10.1200/jco.2015.65.8427] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Don S Dizon
- Don S. Dizon, Massachusetts General Hospital, Boston; Erica Mayer, Dana-Farber Cancer Institute, Boston, MA; Lada Krilov and Richard Schilsky, American Society of Clinical Oncology, Alexandria, VA; Ezra Cohen, University of California San Diego, San Diego; Patricia A. Ganz, University of California Los Angeles, Los Angeles; Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte, CA; Tara Gangadhar, University of Pennsylvania; Stephen Hunger, Children's Hospital Center of Philadelphia, Philadelphia, PA; Thomas A. Hensing, University of Chicago, Evanston, IL; Smitha S. Krishnamurthi, Case Western Reserve University, Cleveland, OH; Andrew B. Lassman and Gary K. Schwartz, Columbia University; David R. Spriggs, Memorial Sloan Kettering Cancer Center; Gina Villani, Ralph Lauren Center for Cancer Care and Prevention, New York, NY; Merry Jennifer Markham, University of Florida, Gainesville; Miguel Angel Villalona-Calero, Miami Cancer Institute, Miami, FL; Michael Neuss, Vanderbilt-Ingram Cancer Center, Nashville, TN; Lisa C. Richardson, Centers for Disease Control and Prevention, Atlanta, GA; and Gregory Masters, Helen F. Graham Cancer Center and Research Institute, Newark, DE
| | - Lada Krilov
- Don S. Dizon, Massachusetts General Hospital, Boston; Erica Mayer, Dana-Farber Cancer Institute, Boston, MA; Lada Krilov and Richard Schilsky, American Society of Clinical Oncology, Alexandria, VA; Ezra Cohen, University of California San Diego, San Diego; Patricia A. Ganz, University of California Los Angeles, Los Angeles; Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte, CA; Tara Gangadhar, University of Pennsylvania; Stephen Hunger, Children's Hospital Center of Philadelphia, Philadelphia, PA; Thomas A. Hensing, University of Chicago, Evanston, IL; Smitha S. Krishnamurthi, Case Western Reserve University, Cleveland, OH; Andrew B. Lassman and Gary K. Schwartz, Columbia University; David R. Spriggs, Memorial Sloan Kettering Cancer Center; Gina Villani, Ralph Lauren Center for Cancer Care and Prevention, New York, NY; Merry Jennifer Markham, University of Florida, Gainesville; Miguel Angel Villalona-Calero, Miami Cancer Institute, Miami, FL; Michael Neuss, Vanderbilt-Ingram Cancer Center, Nashville, TN; Lisa C. Richardson, Centers for Disease Control and Prevention, Atlanta, GA; and Gregory Masters, Helen F. Graham Cancer Center and Research Institute, Newark, DE.
| | - Ezra Cohen
- Don S. Dizon, Massachusetts General Hospital, Boston; Erica Mayer, Dana-Farber Cancer Institute, Boston, MA; Lada Krilov and Richard Schilsky, American Society of Clinical Oncology, Alexandria, VA; Ezra Cohen, University of California San Diego, San Diego; Patricia A. Ganz, University of California Los Angeles, Los Angeles; Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte, CA; Tara Gangadhar, University of Pennsylvania; Stephen Hunger, Children's Hospital Center of Philadelphia, Philadelphia, PA; Thomas A. Hensing, University of Chicago, Evanston, IL; Smitha S. Krishnamurthi, Case Western Reserve University, Cleveland, OH; Andrew B. Lassman and Gary K. Schwartz, Columbia University; David R. Spriggs, Memorial Sloan Kettering Cancer Center; Gina Villani, Ralph Lauren Center for Cancer Care and Prevention, New York, NY; Merry Jennifer Markham, University of Florida, Gainesville; Miguel Angel Villalona-Calero, Miami Cancer Institute, Miami, FL; Michael Neuss, Vanderbilt-Ingram Cancer Center, Nashville, TN; Lisa C. Richardson, Centers for Disease Control and Prevention, Atlanta, GA; and Gregory Masters, Helen F. Graham Cancer Center and Research Institute, Newark, DE
| | - Tara Gangadhar
- Don S. Dizon, Massachusetts General Hospital, Boston; Erica Mayer, Dana-Farber Cancer Institute, Boston, MA; Lada Krilov and Richard Schilsky, American Society of Clinical Oncology, Alexandria, VA; Ezra Cohen, University of California San Diego, San Diego; Patricia A. Ganz, University of California Los Angeles, Los Angeles; Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte, CA; Tara Gangadhar, University of Pennsylvania; Stephen Hunger, Children's Hospital Center of Philadelphia, Philadelphia, PA; Thomas A. Hensing, University of Chicago, Evanston, IL; Smitha S. Krishnamurthi, Case Western Reserve University, Cleveland, OH; Andrew B. Lassman and Gary K. Schwartz, Columbia University; David R. Spriggs, Memorial Sloan Kettering Cancer Center; Gina Villani, Ralph Lauren Center for Cancer Care and Prevention, New York, NY; Merry Jennifer Markham, University of Florida, Gainesville; Miguel Angel Villalona-Calero, Miami Cancer Institute, Miami, FL; Michael Neuss, Vanderbilt-Ingram Cancer Center, Nashville, TN; Lisa C. Richardson, Centers for Disease Control and Prevention, Atlanta, GA; and Gregory Masters, Helen F. Graham Cancer Center and Research Institute, Newark, DE
| | - Patricia A Ganz
- Don S. Dizon, Massachusetts General Hospital, Boston; Erica Mayer, Dana-Farber Cancer Institute, Boston, MA; Lada Krilov and Richard Schilsky, American Society of Clinical Oncology, Alexandria, VA; Ezra Cohen, University of California San Diego, San Diego; Patricia A. Ganz, University of California Los Angeles, Los Angeles; Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte, CA; Tara Gangadhar, University of Pennsylvania; Stephen Hunger, Children's Hospital Center of Philadelphia, Philadelphia, PA; Thomas A. Hensing, University of Chicago, Evanston, IL; Smitha S. Krishnamurthi, Case Western Reserve University, Cleveland, OH; Andrew B. Lassman and Gary K. Schwartz, Columbia University; David R. Spriggs, Memorial Sloan Kettering Cancer Center; Gina Villani, Ralph Lauren Center for Cancer Care and Prevention, New York, NY; Merry Jennifer Markham, University of Florida, Gainesville; Miguel Angel Villalona-Calero, Miami Cancer Institute, Miami, FL; Michael Neuss, Vanderbilt-Ingram Cancer Center, Nashville, TN; Lisa C. Richardson, Centers for Disease Control and Prevention, Atlanta, GA; and Gregory Masters, Helen F. Graham Cancer Center and Research Institute, Newark, DE
| | - Thomas A Hensing
- Don S. Dizon, Massachusetts General Hospital, Boston; Erica Mayer, Dana-Farber Cancer Institute, Boston, MA; Lada Krilov and Richard Schilsky, American Society of Clinical Oncology, Alexandria, VA; Ezra Cohen, University of California San Diego, San Diego; Patricia A. Ganz, University of California Los Angeles, Los Angeles; Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte, CA; Tara Gangadhar, University of Pennsylvania; Stephen Hunger, Children's Hospital Center of Philadelphia, Philadelphia, PA; Thomas A. Hensing, University of Chicago, Evanston, IL; Smitha S. Krishnamurthi, Case Western Reserve University, Cleveland, OH; Andrew B. Lassman and Gary K. Schwartz, Columbia University; David R. Spriggs, Memorial Sloan Kettering Cancer Center; Gina Villani, Ralph Lauren Center for Cancer Care and Prevention, New York, NY; Merry Jennifer Markham, University of Florida, Gainesville; Miguel Angel Villalona-Calero, Miami Cancer Institute, Miami, FL; Michael Neuss, Vanderbilt-Ingram Cancer Center, Nashville, TN; Lisa C. Richardson, Centers for Disease Control and Prevention, Atlanta, GA; and Gregory Masters, Helen F. Graham Cancer Center and Research Institute, Newark, DE
| | - Stephen Hunger
- Don S. Dizon, Massachusetts General Hospital, Boston; Erica Mayer, Dana-Farber Cancer Institute, Boston, MA; Lada Krilov and Richard Schilsky, American Society of Clinical Oncology, Alexandria, VA; Ezra Cohen, University of California San Diego, San Diego; Patricia A. Ganz, University of California Los Angeles, Los Angeles; Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte, CA; Tara Gangadhar, University of Pennsylvania; Stephen Hunger, Children's Hospital Center of Philadelphia, Philadelphia, PA; Thomas A. Hensing, University of Chicago, Evanston, IL; Smitha S. Krishnamurthi, Case Western Reserve University, Cleveland, OH; Andrew B. Lassman and Gary K. Schwartz, Columbia University; David R. Spriggs, Memorial Sloan Kettering Cancer Center; Gina Villani, Ralph Lauren Center for Cancer Care and Prevention, New York, NY; Merry Jennifer Markham, University of Florida, Gainesville; Miguel Angel Villalona-Calero, Miami Cancer Institute, Miami, FL; Michael Neuss, Vanderbilt-Ingram Cancer Center, Nashville, TN; Lisa C. Richardson, Centers for Disease Control and Prevention, Atlanta, GA; and Gregory Masters, Helen F. Graham Cancer Center and Research Institute, Newark, DE
| | - Smitha S Krishnamurthi
- Don S. Dizon, Massachusetts General Hospital, Boston; Erica Mayer, Dana-Farber Cancer Institute, Boston, MA; Lada Krilov and Richard Schilsky, American Society of Clinical Oncology, Alexandria, VA; Ezra Cohen, University of California San Diego, San Diego; Patricia A. Ganz, University of California Los Angeles, Los Angeles; Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte, CA; Tara Gangadhar, University of Pennsylvania; Stephen Hunger, Children's Hospital Center of Philadelphia, Philadelphia, PA; Thomas A. Hensing, University of Chicago, Evanston, IL; Smitha S. Krishnamurthi, Case Western Reserve University, Cleveland, OH; Andrew B. Lassman and Gary K. Schwartz, Columbia University; David R. Spriggs, Memorial Sloan Kettering Cancer Center; Gina Villani, Ralph Lauren Center for Cancer Care and Prevention, New York, NY; Merry Jennifer Markham, University of Florida, Gainesville; Miguel Angel Villalona-Calero, Miami Cancer Institute, Miami, FL; Michael Neuss, Vanderbilt-Ingram Cancer Center, Nashville, TN; Lisa C. Richardson, Centers for Disease Control and Prevention, Atlanta, GA; and Gregory Masters, Helen F. Graham Cancer Center and Research Institute, Newark, DE
| | - Andrew B Lassman
- Don S. Dizon, Massachusetts General Hospital, Boston; Erica Mayer, Dana-Farber Cancer Institute, Boston, MA; Lada Krilov and Richard Schilsky, American Society of Clinical Oncology, Alexandria, VA; Ezra Cohen, University of California San Diego, San Diego; Patricia A. Ganz, University of California Los Angeles, Los Angeles; Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte, CA; Tara Gangadhar, University of Pennsylvania; Stephen Hunger, Children's Hospital Center of Philadelphia, Philadelphia, PA; Thomas A. Hensing, University of Chicago, Evanston, IL; Smitha S. Krishnamurthi, Case Western Reserve University, Cleveland, OH; Andrew B. Lassman and Gary K. Schwartz, Columbia University; David R. Spriggs, Memorial Sloan Kettering Cancer Center; Gina Villani, Ralph Lauren Center for Cancer Care and Prevention, New York, NY; Merry Jennifer Markham, University of Florida, Gainesville; Miguel Angel Villalona-Calero, Miami Cancer Institute, Miami, FL; Michael Neuss, Vanderbilt-Ingram Cancer Center, Nashville, TN; Lisa C. Richardson, Centers for Disease Control and Prevention, Atlanta, GA; and Gregory Masters, Helen F. Graham Cancer Center and Research Institute, Newark, DE
| | - Merry Jennifer Markham
- Don S. Dizon, Massachusetts General Hospital, Boston; Erica Mayer, Dana-Farber Cancer Institute, Boston, MA; Lada Krilov and Richard Schilsky, American Society of Clinical Oncology, Alexandria, VA; Ezra Cohen, University of California San Diego, San Diego; Patricia A. Ganz, University of California Los Angeles, Los Angeles; Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte, CA; Tara Gangadhar, University of Pennsylvania; Stephen Hunger, Children's Hospital Center of Philadelphia, Philadelphia, PA; Thomas A. Hensing, University of Chicago, Evanston, IL; Smitha S. Krishnamurthi, Case Western Reserve University, Cleveland, OH; Andrew B. Lassman and Gary K. Schwartz, Columbia University; David R. Spriggs, Memorial Sloan Kettering Cancer Center; Gina Villani, Ralph Lauren Center for Cancer Care and Prevention, New York, NY; Merry Jennifer Markham, University of Florida, Gainesville; Miguel Angel Villalona-Calero, Miami Cancer Institute, Miami, FL; Michael Neuss, Vanderbilt-Ingram Cancer Center, Nashville, TN; Lisa C. Richardson, Centers for Disease Control and Prevention, Atlanta, GA; and Gregory Masters, Helen F. Graham Cancer Center and Research Institute, Newark, DE
| | - Erica Mayer
- Don S. Dizon, Massachusetts General Hospital, Boston; Erica Mayer, Dana-Farber Cancer Institute, Boston, MA; Lada Krilov and Richard Schilsky, American Society of Clinical Oncology, Alexandria, VA; Ezra Cohen, University of California San Diego, San Diego; Patricia A. Ganz, University of California Los Angeles, Los Angeles; Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte, CA; Tara Gangadhar, University of Pennsylvania; Stephen Hunger, Children's Hospital Center of Philadelphia, Philadelphia, PA; Thomas A. Hensing, University of Chicago, Evanston, IL; Smitha S. Krishnamurthi, Case Western Reserve University, Cleveland, OH; Andrew B. Lassman and Gary K. Schwartz, Columbia University; David R. Spriggs, Memorial Sloan Kettering Cancer Center; Gina Villani, Ralph Lauren Center for Cancer Care and Prevention, New York, NY; Merry Jennifer Markham, University of Florida, Gainesville; Miguel Angel Villalona-Calero, Miami Cancer Institute, Miami, FL; Michael Neuss, Vanderbilt-Ingram Cancer Center, Nashville, TN; Lisa C. Richardson, Centers for Disease Control and Prevention, Atlanta, GA; and Gregory Masters, Helen F. Graham Cancer Center and Research Institute, Newark, DE
| | - Michael Neuss
- Don S. Dizon, Massachusetts General Hospital, Boston; Erica Mayer, Dana-Farber Cancer Institute, Boston, MA; Lada Krilov and Richard Schilsky, American Society of Clinical Oncology, Alexandria, VA; Ezra Cohen, University of California San Diego, San Diego; Patricia A. Ganz, University of California Los Angeles, Los Angeles; Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte, CA; Tara Gangadhar, University of Pennsylvania; Stephen Hunger, Children's Hospital Center of Philadelphia, Philadelphia, PA; Thomas A. Hensing, University of Chicago, Evanston, IL; Smitha S. Krishnamurthi, Case Western Reserve University, Cleveland, OH; Andrew B. Lassman and Gary K. Schwartz, Columbia University; David R. Spriggs, Memorial Sloan Kettering Cancer Center; Gina Villani, Ralph Lauren Center for Cancer Care and Prevention, New York, NY; Merry Jennifer Markham, University of Florida, Gainesville; Miguel Angel Villalona-Calero, Miami Cancer Institute, Miami, FL; Michael Neuss, Vanderbilt-Ingram Cancer Center, Nashville, TN; Lisa C. Richardson, Centers for Disease Control and Prevention, Atlanta, GA; and Gregory Masters, Helen F. Graham Cancer Center and Research Institute, Newark, DE
| | - Sumanta Kumar Pal
- Don S. Dizon, Massachusetts General Hospital, Boston; Erica Mayer, Dana-Farber Cancer Institute, Boston, MA; Lada Krilov and Richard Schilsky, American Society of Clinical Oncology, Alexandria, VA; Ezra Cohen, University of California San Diego, San Diego; Patricia A. Ganz, University of California Los Angeles, Los Angeles; Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte, CA; Tara Gangadhar, University of Pennsylvania; Stephen Hunger, Children's Hospital Center of Philadelphia, Philadelphia, PA; Thomas A. Hensing, University of Chicago, Evanston, IL; Smitha S. Krishnamurthi, Case Western Reserve University, Cleveland, OH; Andrew B. Lassman and Gary K. Schwartz, Columbia University; David R. Spriggs, Memorial Sloan Kettering Cancer Center; Gina Villani, Ralph Lauren Center for Cancer Care and Prevention, New York, NY; Merry Jennifer Markham, University of Florida, Gainesville; Miguel Angel Villalona-Calero, Miami Cancer Institute, Miami, FL; Michael Neuss, Vanderbilt-Ingram Cancer Center, Nashville, TN; Lisa C. Richardson, Centers for Disease Control and Prevention, Atlanta, GA; and Gregory Masters, Helen F. Graham Cancer Center and Research Institute, Newark, DE
| | - Lisa C Richardson
- Don S. Dizon, Massachusetts General Hospital, Boston; Erica Mayer, Dana-Farber Cancer Institute, Boston, MA; Lada Krilov and Richard Schilsky, American Society of Clinical Oncology, Alexandria, VA; Ezra Cohen, University of California San Diego, San Diego; Patricia A. Ganz, University of California Los Angeles, Los Angeles; Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte, CA; Tara Gangadhar, University of Pennsylvania; Stephen Hunger, Children's Hospital Center of Philadelphia, Philadelphia, PA; Thomas A. Hensing, University of Chicago, Evanston, IL; Smitha S. Krishnamurthi, Case Western Reserve University, Cleveland, OH; Andrew B. Lassman and Gary K. Schwartz, Columbia University; David R. Spriggs, Memorial Sloan Kettering Cancer Center; Gina Villani, Ralph Lauren Center for Cancer Care and Prevention, New York, NY; Merry Jennifer Markham, University of Florida, Gainesville; Miguel Angel Villalona-Calero, Miami Cancer Institute, Miami, FL; Michael Neuss, Vanderbilt-Ingram Cancer Center, Nashville, TN; Lisa C. Richardson, Centers for Disease Control and Prevention, Atlanta, GA; and Gregory Masters, Helen F. Graham Cancer Center and Research Institute, Newark, DE
| | - Richard Schilsky
- Don S. Dizon, Massachusetts General Hospital, Boston; Erica Mayer, Dana-Farber Cancer Institute, Boston, MA; Lada Krilov and Richard Schilsky, American Society of Clinical Oncology, Alexandria, VA; Ezra Cohen, University of California San Diego, San Diego; Patricia A. Ganz, University of California Los Angeles, Los Angeles; Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte, CA; Tara Gangadhar, University of Pennsylvania; Stephen Hunger, Children's Hospital Center of Philadelphia, Philadelphia, PA; Thomas A. Hensing, University of Chicago, Evanston, IL; Smitha S. Krishnamurthi, Case Western Reserve University, Cleveland, OH; Andrew B. Lassman and Gary K. Schwartz, Columbia University; David R. Spriggs, Memorial Sloan Kettering Cancer Center; Gina Villani, Ralph Lauren Center for Cancer Care and Prevention, New York, NY; Merry Jennifer Markham, University of Florida, Gainesville; Miguel Angel Villalona-Calero, Miami Cancer Institute, Miami, FL; Michael Neuss, Vanderbilt-Ingram Cancer Center, Nashville, TN; Lisa C. Richardson, Centers for Disease Control and Prevention, Atlanta, GA; and Gregory Masters, Helen F. Graham Cancer Center and Research Institute, Newark, DE
| | - Gary K Schwartz
- Don S. Dizon, Massachusetts General Hospital, Boston; Erica Mayer, Dana-Farber Cancer Institute, Boston, MA; Lada Krilov and Richard Schilsky, American Society of Clinical Oncology, Alexandria, VA; Ezra Cohen, University of California San Diego, San Diego; Patricia A. Ganz, University of California Los Angeles, Los Angeles; Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte, CA; Tara Gangadhar, University of Pennsylvania; Stephen Hunger, Children's Hospital Center of Philadelphia, Philadelphia, PA; Thomas A. Hensing, University of Chicago, Evanston, IL; Smitha S. Krishnamurthi, Case Western Reserve University, Cleveland, OH; Andrew B. Lassman and Gary K. Schwartz, Columbia University; David R. Spriggs, Memorial Sloan Kettering Cancer Center; Gina Villani, Ralph Lauren Center for Cancer Care and Prevention, New York, NY; Merry Jennifer Markham, University of Florida, Gainesville; Miguel Angel Villalona-Calero, Miami Cancer Institute, Miami, FL; Michael Neuss, Vanderbilt-Ingram Cancer Center, Nashville, TN; Lisa C. Richardson, Centers for Disease Control and Prevention, Atlanta, GA; and Gregory Masters, Helen F. Graham Cancer Center and Research Institute, Newark, DE
| | - David R Spriggs
- Don S. Dizon, Massachusetts General Hospital, Boston; Erica Mayer, Dana-Farber Cancer Institute, Boston, MA; Lada Krilov and Richard Schilsky, American Society of Clinical Oncology, Alexandria, VA; Ezra Cohen, University of California San Diego, San Diego; Patricia A. Ganz, University of California Los Angeles, Los Angeles; Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte, CA; Tara Gangadhar, University of Pennsylvania; Stephen Hunger, Children's Hospital Center of Philadelphia, Philadelphia, PA; Thomas A. Hensing, University of Chicago, Evanston, IL; Smitha S. Krishnamurthi, Case Western Reserve University, Cleveland, OH; Andrew B. Lassman and Gary K. Schwartz, Columbia University; David R. Spriggs, Memorial Sloan Kettering Cancer Center; Gina Villani, Ralph Lauren Center for Cancer Care and Prevention, New York, NY; Merry Jennifer Markham, University of Florida, Gainesville; Miguel Angel Villalona-Calero, Miami Cancer Institute, Miami, FL; Michael Neuss, Vanderbilt-Ingram Cancer Center, Nashville, TN; Lisa C. Richardson, Centers for Disease Control and Prevention, Atlanta, GA; and Gregory Masters, Helen F. Graham Cancer Center and Research Institute, Newark, DE
| | - Miguel Angel Villalona-Calero
- Don S. Dizon, Massachusetts General Hospital, Boston; Erica Mayer, Dana-Farber Cancer Institute, Boston, MA; Lada Krilov and Richard Schilsky, American Society of Clinical Oncology, Alexandria, VA; Ezra Cohen, University of California San Diego, San Diego; Patricia A. Ganz, University of California Los Angeles, Los Angeles; Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte, CA; Tara Gangadhar, University of Pennsylvania; Stephen Hunger, Children's Hospital Center of Philadelphia, Philadelphia, PA; Thomas A. Hensing, University of Chicago, Evanston, IL; Smitha S. Krishnamurthi, Case Western Reserve University, Cleveland, OH; Andrew B. Lassman and Gary K. Schwartz, Columbia University; David R. Spriggs, Memorial Sloan Kettering Cancer Center; Gina Villani, Ralph Lauren Center for Cancer Care and Prevention, New York, NY; Merry Jennifer Markham, University of Florida, Gainesville; Miguel Angel Villalona-Calero, Miami Cancer Institute, Miami, FL; Michael Neuss, Vanderbilt-Ingram Cancer Center, Nashville, TN; Lisa C. Richardson, Centers for Disease Control and Prevention, Atlanta, GA; and Gregory Masters, Helen F. Graham Cancer Center and Research Institute, Newark, DE
| | - Gina Villani
- Don S. Dizon, Massachusetts General Hospital, Boston; Erica Mayer, Dana-Farber Cancer Institute, Boston, MA; Lada Krilov and Richard Schilsky, American Society of Clinical Oncology, Alexandria, VA; Ezra Cohen, University of California San Diego, San Diego; Patricia A. Ganz, University of California Los Angeles, Los Angeles; Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte, CA; Tara Gangadhar, University of Pennsylvania; Stephen Hunger, Children's Hospital Center of Philadelphia, Philadelphia, PA; Thomas A. Hensing, University of Chicago, Evanston, IL; Smitha S. Krishnamurthi, Case Western Reserve University, Cleveland, OH; Andrew B. Lassman and Gary K. Schwartz, Columbia University; David R. Spriggs, Memorial Sloan Kettering Cancer Center; Gina Villani, Ralph Lauren Center for Cancer Care and Prevention, New York, NY; Merry Jennifer Markham, University of Florida, Gainesville; Miguel Angel Villalona-Calero, Miami Cancer Institute, Miami, FL; Michael Neuss, Vanderbilt-Ingram Cancer Center, Nashville, TN; Lisa C. Richardson, Centers for Disease Control and Prevention, Atlanta, GA; and Gregory Masters, Helen F. Graham Cancer Center and Research Institute, Newark, DE
| | - Gregory Masters
- Don S. Dizon, Massachusetts General Hospital, Boston; Erica Mayer, Dana-Farber Cancer Institute, Boston, MA; Lada Krilov and Richard Schilsky, American Society of Clinical Oncology, Alexandria, VA; Ezra Cohen, University of California San Diego, San Diego; Patricia A. Ganz, University of California Los Angeles, Los Angeles; Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte, CA; Tara Gangadhar, University of Pennsylvania; Stephen Hunger, Children's Hospital Center of Philadelphia, Philadelphia, PA; Thomas A. Hensing, University of Chicago, Evanston, IL; Smitha S. Krishnamurthi, Case Western Reserve University, Cleveland, OH; Andrew B. Lassman and Gary K. Schwartz, Columbia University; David R. Spriggs, Memorial Sloan Kettering Cancer Center; Gina Villani, Ralph Lauren Center for Cancer Care and Prevention, New York, NY; Merry Jennifer Markham, University of Florida, Gainesville; Miguel Angel Villalona-Calero, Miami Cancer Institute, Miami, FL; Michael Neuss, Vanderbilt-Ingram Cancer Center, Nashville, TN; Lisa C. Richardson, Centers for Disease Control and Prevention, Atlanta, GA; and Gregory Masters, Helen F. Graham Cancer Center and Research Institute, Newark, DE
| |
Collapse
|
5929
|
Jin C, Zhang X, Zhao K, Xu J, Zhao M, Xu X. The efficacy and safety of nivolumab in the treatment of advanced melanoma: a meta-analysis of clinical trials. Onco Targets Ther 2016; 9:1571-8. [PMID: 27051297 PMCID: PMC4803248 DOI: 10.2147/ott.s96762] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background Nivolumab has become a therapeutic regimen for the treatment of patients with advanced melanoma. The goal of this study was to assess the efficacy and safety of nivolumab in patients with advanced melanoma. Methods A systematic search from January 2008 to August 2015 with “nivolumab” and “advanced melanoma” as search terms was performed for possible clinical trials. According to the hazard ratio and the 95% confidence interval (CI) for progression-free survival (PFS), rates of objective response, complete response, partial response, rates of toxic effects, and the efficacy and safety of nivolumab were assessed. Using the software Review Manager (version 5.3) a meta-analysis was performed. Results There were four trials with 1,910 patients included. Based on the four trials, the pooled hazard ratio of PFS was 0.53 (95% CI, 0.43–0.66; P<0.001). The pooled risk ratio for the objective response rate, complete response, and partial response was 2.98% (95% CI, 2.38%–3.73%; P<0.001), 3.71% (95% CI, 2.67%–5.14%; P<0.001), and 2.51% (95% CI, 2.12%–2.99%; P<0.001), respectively. Nivolumab plus ipilimumab therapy significantly increased the risk of grade 3/4 rash and fatigue. Conclusion Nivolumab-based therapy prolonged PFS in treatment of advanced melanoma, with less adverse effects. Nivolumab appears to be a favorable treatment option as a novel, targeted anticancer agent, for patients with advanced melanoma.
Collapse
Affiliation(s)
- Conghui Jin
- Department of Medical Oncology, Nantong Tumor Hospital, Nantong, People's Republic of China
| | - Xunlei Zhang
- Department of Medical Oncology, Nantong Tumor Hospital, Nantong, People's Republic of China
| | - Kuiling Zhao
- Department of Medical Oncology, Nantong Tumor Hospital, Nantong, People's Republic of China
| | - Jun Xu
- Department of Medical Oncology, Nantong Tumor Hospital, Nantong, People's Republic of China
| | - Min Zhao
- Department of Medical Oncology, Nantong Tumor Hospital, Nantong, People's Republic of China
| | - Xiaohong Xu
- Department of Medical Oncology, Nantong Tumor Hospital, Nantong, People's Republic of China
| |
Collapse
|
5930
|
Ladoire S, Senovilla L, Enot D, Ghiringhelli F, Poirier-Colame V, Chaba K, Erdag G, Schaefer JT, Deacon DH, Zitvogel L, Slingluff CL, Kroemer G. Biomarkers of immunogenic stress in metastases from melanoma patients: Correlations with the immune infiltrate. Oncoimmunology 2016; 5:e1160193. [PMID: 27471635 DOI: 10.1080/2162402x.2016.1160193] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 02/24/2016] [Accepted: 02/25/2016] [Indexed: 01/21/2023] Open
Abstract
Melanoma is known to be under latent immunosurveillance. Here, we studied four biomarkers of immunogenic cell stress and death (microtubule-associated proteins 1A/1B light chain 3B (MAP-LC3B, best known as LC3B)-positive puncta in the cytoplasm as a sign of autophagy; presence of nuclear HMGB1; phosphorylation of eIF2α; increase in ploidy) in melanoma cells, in tissue microarrays (TMA) from metastases from 147 melanoma patients. These biomarkers of immunogenicity were correlated with the density of immune cells infiltrating the metastases and expressing CD3, CD4(+), CD8(+), CD20, CD45, CD56, CD138, CD163, DC-LAMP or FOXP3. LC3B puncta positively correlated with the infiltration of metastases by CD163(+) macrophages, while expression of HMGB1 correlated with infiltration by FOXP3(+) regulatory T cells and CD56(+) lymphocytes. eIF2α phosphorylation was associated with an augmentation of nuclear diameters, reflecting an increase in ploidy. Interestingly, therapeutic vaccination led to a reduction of eIF2α phosphorylation suggestive of immunoselection against cells bearing this sign of endoplasmic reticulum (ER) stress. None of the stress/death-related biomarkers had a significant prognostic impact, contrasting with the major prognostic effect of the ratio of cytotoxic T lymphocytes (CTL) over immunosuppressive FOXP3(+) and CD163(+) cells. Altogether, these results support the idea of a mutual dialog between, on one hand, melanoma cells with their cell-intrinsic stress pathways and, on the other hand, immune effectors. Future work is required to understand the detailed mechanisms of this interaction.
Collapse
Affiliation(s)
- Sylvain Ladoire
- Department of Medical Oncology, Georges François Leclerc Center, Dijon, France; Institut National de la Santé et de la Recherche Médicale, Avenir Team INSERM, University of Burgundy, Dijon, France; Institut National de la Santé et de la Recherche Médicale, U1015, Equipe labellisée Ligue Nationale Contre le Cancer, Institut Gustave Roussy, Villejuif, France
| | - Laura Senovilla
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France; INSERM, U1138, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; University of Paris Sud XI, Villejuif, France
| | - David Enot
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus , Villejuif, France
| | - François Ghiringhelli
- Department of Medical Oncology, Georges François Leclerc Center, Dijon, France; Institut National de la Santé et de la Recherche Médicale, Avenir Team INSERM, University of Burgundy, Dijon, France
| | - Vichnou Poirier-Colame
- Institut National de la Santé et de la Recherche Médicale, U1015, Equipe labellisée Ligue Nationale Contre le Cancer, Institut Gustave Roussy , Villejuif, France
| | - Kariman Chaba
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France; INSERM, U1138, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Gulsun Erdag
- Department of Pathology, University of Virginia, Charlottesville, VA, USA; Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Jochen T Schaefer
- Department of Pathology, University of Virginia, Charlottesville, VA, USA; Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Donna H Deacon
- Department of Surgery, University of Virginia , Charlottesville, VA, USA
| | - Laurence Zitvogel
- Institut National de la Santé et de la Recherche Médicale, U1015, Equipe labellisée Ligue Nationale Contre le Cancer, Institut Gustave Roussy, Villejuif, France; University of Paris Sud XI, Villejuif, France; Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France
| | - Craig L Slingluff
- Department of Surgery, University of Virginia , Charlottesville, VA, USA
| | - Guido Kroemer
- Equipe 11 labellisée pas la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France; INSERM, U1138, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France; Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
5931
|
Loi S, Dushyanthen S, Beavis PA, Salgado R, Denkert C, Savas P, Combs S, Rimm DL, Giltnane JM, Estrada MV, Sánchez V, Sanders ME, Cook RS, Pilkinton MA, Mallal SA, Wang K, Miller VA, Stephens PJ, Yelensky R, Doimi FD, Gómez H, Ryzhov SV, Darcy PK, Arteaga CL, Balko JM. RAS/MAPK Activation Is Associated with Reduced Tumor-Infiltrating Lymphocytes in Triple-Negative Breast Cancer: Therapeutic Cooperation Between MEK and PD-1/PD-L1 Immune Checkpoint Inhibitors. Clin Cancer Res 2016; 22:1499-509. [PMID: 26515496 PMCID: PMC4794351 DOI: 10.1158/1078-0432.ccr-15-1125] [Citation(s) in RCA: 411] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 10/21/2015] [Indexed: 02/07/2023]
Abstract
PURPOSE Tumor-infiltrating lymphocytes (TIL) in the residual disease (RD) of triple-negative breast cancers (TNBC) after neoadjuvant chemotherapy (NAC) are associated with improved survival, but insight into tumor cell-autonomous molecular pathways affecting these features are lacking. EXPERIMENTAL DESIGN We analyzed TILs in the RD of clinically and molecularly characterized TNBCs after NAC and explored therapeutic strategies targeting combinations of MEK inhibitors with PD-1/PD-L1-targeted immunotherapy in mouse models of breast cancer. RESULTS Presence of TILs in the RD was significantly associated with improved prognosis. Genetic or transcriptomic alterations in Ras-MAPK signaling were significantly correlated with lower TILs. MEK inhibition upregulated cell surface MHC expression and PD-L1 in TNBC cells both in vivo and in vitro. Moreover, combined MEK and PD-L1/PD-1 inhibition enhanced antitumor immune responses in mouse models of breast cancer. CONCLUSIONS These data suggest the possibility that Ras-MAPK pathway activation promotes immune-evasion in TNBC, and support clinical trials combining MEK- and PD-L1-targeted therapies. Furthermore, Ras/MAPK activation and MHC expression may be predictive biomarkers of response to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Sherene Loi
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia.
| | | | - Paul A Beavis
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Roberto Salgado
- Breast Cancer Translational Research Laboratory, Institute Jules Bordet, Brussels, Department of Pathology, GZA Antwerp, Belgium
| | - Carsten Denkert
- Charité University and German Cancer Consortium (DKTK), Berlin, Germany
| | - Peter Savas
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Susan Combs
- Departments of Pathology and Medicine, Yale University, New Haven, Connecticut
| | - David L Rimm
- Departments of Pathology and Medicine, Yale University, New Haven, Connecticut
| | - Jennifer M Giltnane
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee. Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Monica V Estrada
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Violeta Sánchez
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Melinda E Sanders
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee. Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Rebecca S Cook
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee. Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee
| | - Mark A Pilkinton
- Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Simon A Mallal
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee. Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Kai Wang
- Foundation Medicine, Cambridge, Massachusetts
| | | | | | | | - Franco D Doimi
- Instituto Nacional de Enfermedades Neoplásicas (INEN), Lima, Perú
| | - Henry Gómez
- Instituto Nacional de Enfermedades Neoplásicas (INEN), Lima, Perú
| | | | - Phillip K Darcy
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Carlos L Arteaga
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee. Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee. Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Justin M Balko
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee. Department of Medicine, Vanderbilt University, Nashville, Tennessee.
| |
Collapse
|
5932
|
Loi S, Dushyanthen S, Beavis PA, Salgado R, Denkert C, Savas P, Combs S, Rimm DL, Giltnane JM, Estrada MV, Sánchez V, Sanders ME, Cook RS, Pilkinton MA, Mallal SA, Wang K, Miller VA, Stephens PJ, Yelensky R, Doimi FD, Gómez H, Ryzhov SV, Darcy PK, Arteaga CL, Balko JM. RAS/MAPK Activation Is Associated with Reduced Tumor-Infiltrating Lymphocytes in Triple-Negative Breast Cancer: Therapeutic Cooperation Between MEK and PD-1/PD-L1 Immune Checkpoint Inhibitors. Clin Cancer Res 2016. [PMID: 26515496 DOI: 10.1158/1078-0432.ccr-15-1125.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Tumor-infiltrating lymphocytes (TIL) in the residual disease (RD) of triple-negative breast cancers (TNBC) after neoadjuvant chemotherapy (NAC) are associated with improved survival, but insight into tumor cell-autonomous molecular pathways affecting these features are lacking. EXPERIMENTAL DESIGN We analyzed TILs in the RD of clinically and molecularly characterized TNBCs after NAC and explored therapeutic strategies targeting combinations of MEK inhibitors with PD-1/PD-L1-targeted immunotherapy in mouse models of breast cancer. RESULTS Presence of TILs in the RD was significantly associated with improved prognosis. Genetic or transcriptomic alterations in Ras-MAPK signaling were significantly correlated with lower TILs. MEK inhibition upregulated cell surface MHC expression and PD-L1 in TNBC cells both in vivo and in vitro. Moreover, combined MEK and PD-L1/PD-1 inhibition enhanced antitumor immune responses in mouse models of breast cancer. CONCLUSIONS These data suggest the possibility that Ras-MAPK pathway activation promotes immune-evasion in TNBC, and support clinical trials combining MEK- and PD-L1-targeted therapies. Furthermore, Ras/MAPK activation and MHC expression may be predictive biomarkers of response to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Sherene Loi
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia.
| | | | - Paul A Beavis
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Roberto Salgado
- Breast Cancer Translational Research Laboratory, Institute Jules Bordet, Brussels, Department of Pathology, GZA Antwerp, Belgium
| | - Carsten Denkert
- Charité University and German Cancer Consortium (DKTK), Berlin, Germany
| | - Peter Savas
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Susan Combs
- Departments of Pathology and Medicine, Yale University, New Haven, Connecticut
| | - David L Rimm
- Departments of Pathology and Medicine, Yale University, New Haven, Connecticut
| | - Jennifer M Giltnane
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee. Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Monica V Estrada
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Violeta Sánchez
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Melinda E Sanders
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee. Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Rebecca S Cook
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee. Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee
| | - Mark A Pilkinton
- Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Simon A Mallal
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee. Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Kai Wang
- Foundation Medicine, Cambridge, Massachusetts
| | | | | | | | - Franco D Doimi
- Instituto Nacional de Enfermedades Neoplásicas (INEN), Lima, Perú
| | - Henry Gómez
- Instituto Nacional de Enfermedades Neoplásicas (INEN), Lima, Perú
| | | | - Phillip K Darcy
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Carlos L Arteaga
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee. Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee. Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Justin M Balko
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee. Department of Medicine, Vanderbilt University, Nashville, Tennessee.
| |
Collapse
|
5933
|
The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nat Rev Clin Oncol 2016; 13:273-90. [PMID: 26977780 DOI: 10.1038/nrclinonc.2016.25] [Citation(s) in RCA: 782] [Impact Index Per Article: 86.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the past decade, advances in the use of monoclonal antibodies (mAbs) and adoptive cellular therapy to treat cancer by modulating the immune response have led to unprecedented responses in patients with advanced-stage tumours that would otherwise have been fatal. To date, three immune-checkpoint-blocking mAbs have been approved in the USA for the treatment of patients with several types of cancer, and more patients will benefit from immunomodulatory mAb therapy in the months and years ahead. Concurrently, the adoptive transfer of genetically modified lymphocytes to treat patients with haematological malignancies has yielded dramatic results, and we anticipate that this approach will rapidly become the standard of care for an increasing number of patients. In this Review, we highlight the latest advances in immunotherapy and discuss the role that it will have in the future of cancer treatment, including settings for which testing combination strategies and 'armoured' CAR T cells are recommended.
Collapse
|
5934
|
Bell RB, Leidner R, Feng Z, Crittenden MR, Gough MJ, Fox BA. Developing an Immunotherapy Strategy for the Effective Treatment of Oral, Head and Neck Squamous Cell Carcinoma. J Oral Maxillofac Surg 2016; 73:S107-15. [PMID: 26608138 DOI: 10.1016/j.joms.2015.05.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/22/2015] [Indexed: 12/12/2022]
Affiliation(s)
- R Bryan Bell
- Medical Director, Providence Oral, Head and Neck Cancer Program and Clinic, Providence Cancer Center; Investigator, Robert W. Franz Cancer Research Center in the Earle A. Chiles Research Institute at Providence Cancer Center; Consultant, The Head and Neck Institute, Portland, OR.
| | - Rom Leidner
- Co-Director, Providence Oral, Head and Neck Cancer Program, Providence Cancer Center; Investigator, Robert W. Franz Cancer Research Center in the Earle A. Chiles Research Institute at Providence Cancer Center, Portland, OR
| | - Zipei Feng
- Predoctoral student, Robert W. Franz Cancer Research Center in the Earle A. Chiles Research Institute at Providence Cancer Center; MD/PhD Program, Oregon Health and Science University, Portland, OR
| | - Marka R Crittenden
- Director of Translational Radiation Research, Robert W. Franz Cancer Research Center in the Earle A. Chiles Research Institute at Providence Cancer Center; Radiation Oncologist, Providence Oral, Head and Neck Cancer Program and Clinic, Providence Cancer Center, Portland, OR
| | - Michael J Gough
- Member and Investigator, Robert W. Franz Cancer Research Center in the Earle A. Chiles Research Institute at Providence Cancer Center, Portland, OR
| | - Bernard A Fox
- Chief, Laboratory of Molecular and Tumor Immunology, Robert W. Franz Cancer Research Center in the Earle A. Chiles Research Institute at Providence Cancer Center, Portland, OR
| |
Collapse
|
5935
|
Ren R, Koti M, Hamilton T, Graham CH, Nayak JG, Singh J, Drachenberg DE, Siemens DR. A primer on tumour immunology and prostate cancer immunotherapy. Can Urol Assoc J 2016; 10:60-5. [PMID: 26977209 DOI: 10.5489/cuaj.3418] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
No abstract
Collapse
Affiliation(s)
- Runhan Ren
- Department of Urology, Queen's University, Kingston, ON, Canada
| | - Madhuri Koti
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Thomas Hamilton
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Charles H Graham
- Department of Urology, Queen's University, Kingston, ON, Canada;; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Jasmir G Nayak
- Section of Urology, University of Manitoba, Winnipeg, MB, Canada
| | - Jas Singh
- Section of Urology, University of Manitoba, Winnipeg, MB, Canada
| | | | - D Robert Siemens
- Department of Urology, Queen's University, Kingston, ON, Canada;; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| |
Collapse
|
5936
|
Slimano F, Roessle C, Blanc C, De Maleissye MF, Bauler S. [Updates on prevention and treatment of melanoma: Pharmacist involvements and challenges]. ANNALES PHARMACEUTIQUES FRANÇAISES 2016; 74:335-49. [PMID: 26968263 DOI: 10.1016/j.pharma.2016.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 01/25/2016] [Accepted: 01/27/2016] [Indexed: 02/07/2023]
Abstract
Melanoma is a skin cancer that represents an actual public health problem. Its incidence is increasing every year. Environmental risk factors have been clearly identified. Early diagnosis of a suspicious skin lesion should be possible by any health professionals because the prognosis is correlated with the evolution of the disease and the presence of metastases. The advent of new therapies in metastatic forms with the development of immunotherapies and kinases inhibitors has significantly changed the management of this disease. New therapies are available in retail pharmacies and involve health professionals out of the hospital. This article is intended for community and hospital pharmacists and summarizes recommendations for primary and secondary prevention. It updates on new targeted therapies. It wants to give advices to the community pharmacists about the effective use of those treatments for melanoma.
Collapse
Affiliation(s)
- F Slimano
- Département de pharmacie clinique, Gustave-Roussy cancer campus, 114, rue Édouard-Vaillant, 94805 Villejuif, France; Laboratoire de pharmacologie et pharmacocinétique, UFR de pharmacie, université de Reims-Champagne-Ardenne, 51, rue Cognacq-Jay, 51100 Reims, France; Unité MEDyC, UMR CNRS/URCA, université de Reims-Champagne-Ardenne, 51, rue Cognacq-Jay, 51100 Reims, France.
| | - C Roessle
- Département de pharmacie clinique, Gustave-Roussy cancer campus, 114, rue Édouard-Vaillant, 94805 Villejuif, France
| | - C Blanc
- Service de pharmacie, hôpital Ambroise-Paré, 9, avenue Charles-de-Gaulle, 92104 Boulogne-Billancourt, France
| | - M-F De Maleissye
- Service de dermatologie, hôpital Ambroise-Paré, 9, avenue Charles-de-Gaulle, 92104 Boulogne-Billancourt, France
| | - S Bauler
- Service de pharmacie, hôpital Ambroise-Paré, 9, avenue Charles-de-Gaulle, 92104 Boulogne-Billancourt, France
| |
Collapse
|
5937
|
Hoos A. Development of immuno-oncology drugs - from CTLA4 to PD1 to the next generations. Nat Rev Drug Discov 2016; 15:235-47. [PMID: 26965203 DOI: 10.1038/nrd.2015.35] [Citation(s) in RCA: 460] [Impact Index Per Article: 51.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Since the regulatory approval of ipilimumab in 2011, the field of cancer immunotherapy has been experiencing a renaissance. This success is based on progress in both preclinical and clinical science, including the development of new methods of investigation. Immuno-oncology has become a sub-specialty within oncology owing to its unique science and its potential for substantial and long-term clinical benefit. Immunotherapy agents do not directly attack the tumour but instead mobilize the immune system - this can be achieved through various approaches that utilize adaptive or innate immunity. Therefore, immuno-oncology drug development encompasses a broad range of agents, including antibodies, peptides, proteins, small molecules, adjuvants, cytokines, oncolytic viruses, bi-specific molecules and cellular therapies. This Perspective summarizes the recent history of cancer immunotherapy, including the factors that led to its success, provides an overview of novel drug-development considerations, summarizes three generations of immunotherapies that have been developed since 2011 and, thus, illustrates the breadth of opportunities these new generations of immunotherapies represent.
Collapse
Affiliation(s)
- Axel Hoos
- Oncology Research and Development, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, USA
| |
Collapse
|
5938
|
Zheng H, Zhao W, Yan C, Watson CC, Massengill M, Xie M, Massengill C, Noyes DR, Martinez GV, Afzal R, Chen Z, Ren X, Antonia SJ, Haura EB, Ruffell B, Beg AA. HDAC Inhibitors Enhance T-Cell Chemokine Expression and Augment Response to PD-1 Immunotherapy in Lung Adenocarcinoma. Clin Cancer Res 2016; 22:4119-32. [PMID: 26964571 DOI: 10.1158/1078-0432.ccr-15-2584] [Citation(s) in RCA: 250] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 03/06/2016] [Indexed: 12/24/2022]
Abstract
PURPOSE A significant limitation of checkpoint blockade immunotherapy is the relatively low response rate (e.g., ∼20% with PD-1 blockade in lung cancer). In this study, we tested whether strategies that increase T-cell infiltration to tumors can be efficacious in enhancing immunotherapy response. EXPERIMENTAL DESIGN We performed an unbiased screen to identify FDA-approved oncology agents with an ability to enhance T-cell chemokine expression with the goal of identifying agents capable of augmenting immunotherapy response. Identified agents were tested in multiple lung tumor models as single agents and in combination with PD-1 blockade. Additional molecular and cellular analysis of tumors was used to define underlying mechanisms. RESULTS We found that histone deacetylase (HDAC) inhibitors (HDACi) increased expression of multiple T-cell chemokines in cancer cells, macrophages, and T cells. Using the HDACi romidepsin in vivo, we observed increased chemokine expression, enhanced T-cell infiltration, and T-cell-dependent tumor regression. Importantly, romidepsin significantly enhanced the response to PD-1 blockade immunotherapy in multiple lung tumor models, including nearly complete rejection in two models. Combined romidepsin and PD-1 blockade also significantly enhanced activation of tumor-infiltrating T cells. CONCLUSIONS These results provide evidence for a novel role of HDACs in modulating T-cell chemokine expression in multiple cell types. In addition, our findings indicate that pharmacologic induction of T-cell chemokine expression represents a conceptually novel approach for enhancing immunotherapy response. Finally, these results suggest that combination of HDAC inhibitors with PD-1 blockade represents a promising strategy for lung cancer treatment. Clin Cancer Res; 22(16); 4119-32. ©2016 AACR.
Collapse
Affiliation(s)
- Hong Zheng
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida
| | - Weipeng Zhao
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida. Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Cihui Yan
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida. Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Crystina C Watson
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida. Department of Cancer Biology PhD Program, University of South Florida, Tampa, Florida
| | | | - Mengyu Xie
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida. Department of Cancer Biology PhD Program, University of South Florida, Tampa, Florida
| | | | - David R Noyes
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida
| | - Gary V Martinez
- Department of Cancer Imaging and Metabolism, Moffitt Cancer Center, Tampa, Florida
| | - Roha Afzal
- Department of Cancer Imaging and Metabolism, Moffitt Cancer Center, Tampa, Florida
| | - Zhihua Chen
- Department of Bioinformatics, Moffitt Cancer Center, Tampa, Florida
| | - Xiubao Ren
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Scott J Antonia
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida. Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Eric B Haura
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Brian Ruffell
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida. Department of Breast Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Amer A Beg
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida. Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, Florida.
| |
Collapse
|
5939
|
Madore J, Strbenac D, Vilain R, Menzies AM, Yang JYH, Thompson JF, Long GV, Mann GJ, Scolyer RA, Wilmott JS. PD-L1 Negative Status is Associated with Lower Mutation Burden, Differential Expression of Immune-Related Genes, and Worse Survival in Stage III Melanoma. Clin Cancer Res 2016; 22:3915-23. [PMID: 26960397 DOI: 10.1158/1078-0432.ccr-15-1714] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 02/22/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Understanding why some melanomas test negative for PD-L1 by IHC may have implications for the application of anti-PD-1 therapies in melanoma management. This study sought to determine somatic mutation and gene expression patterns associated with tumor cell PD-L1 expression, or lack thereof, in stage III metastatic melanoma to better define therapeutically relevant patient subgroups. EXPERIMENTAL DESIGN IHC for PD-L1 was assessed in 52 American Joint Committee on Cancer stage III melanoma lymph node specimens and compared with specimen-matched comprehensive clinicopathologic, genomic, and transcriptomic data. RESULTS PD-L1-negative status was associated with lower nonsynonymous mutation (NSM) burden (P = 0.017) and worse melanoma-specific survival [HR = 0.28 (0.12-0.66), P = 0.002] in stage III melanoma. Gene set enrichment analysis identified an immune-related gene expression signature in PD-L1-positive tumors. There was a marked increase in cytotoxic T-cell and macrophage-specific genes in PD-L1-positive melanomas. CD8A(high) gene expression was associated with better melanoma-specific survival [HR = 0.2 (0.05-0.87), P = 0.017] and restricted to PD-L1-positive stage III specimens. NF1 mutations were restricted to PD-L1-positive tumors (P = 0.041). CONCLUSIONS Tumor negative PD-L1 status in stage III melanoma lymph node metastasis is a marker of worse patient survival and is associated with a poor immune response gene signature. Lower NSM levels were associated with PD-L1-negative status suggesting differences in somatic mutation profiles are a determinant of PD-L1-associated antitumor immunity in stage III melanoma. Clin Cancer Res; 22(15); 3915-23. ©2016 AACR.
Collapse
Affiliation(s)
- Jason Madore
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia. Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Dario Strbenac
- School of Mathematics and Statistics, The University of Sydney Camperdown, New South Wales, Australia
| | - Ricardo Vilain
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia. Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia. Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia. Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia. Department of Medical Oncology, Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia
| | - Jeen Y H Yang
- School of Mathematics and Statistics, The University of Sydney Camperdown, New South Wales, Australia
| | - John F Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia. Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia. Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia. Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia. Department of Medical Oncology, Royal North Shore and Mater Hospitals, Sydney, New South Wales, Australia
| | - Graham J Mann
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia. Centre for Cancer Research, The University of Sydney at Westmead Millennium Institute, Westmead, New South Wales, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia. Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia. Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia.
| | - James S Wilmott
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia. Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
5940
|
Spagnolo F, Picasso V, Lambertini M, Ottaviano V, Dozin B, Queirolo P. Survival of patients with metastatic melanoma and brain metastases in the era of MAP-kinase inhibitors and immunologic checkpoint blockade antibodies: A systematic review. Cancer Treat Rev 2016; 45:38-45. [PMID: 26975020 DOI: 10.1016/j.ctrv.2016.03.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 03/01/2016] [Accepted: 03/02/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND The incidence of brain metastases (BM) in melanoma patients is common and associated with poor prognosis. MAP-kinase inhibitors and immunologic checkpoint blockade antibodies led to improved survival of metastatic melanoma patients; however, patients with BM are under-represented or excluded from the majority of clinical trials and the impact of new drugs on their survival is less clear. With the present systematic review, we aimed to analyze outcomes of patients with melanoma BM treated with the new drugs, both in the setting of phase I-II-III clinical trials and in the "real world". METHODS An electronic search was performed to identify studies reporting survival outcomes of patients with melanoma BM treated with MAP-kinase inhibitors and/or immunologic checkpoint blockade antibodies, regardless of study design. RESULTS Twenty-two studies were included for a total of 2153 patients. Median OS was 7.9 months in phase I-II-III trials and 7.7 months in "real world" studies. In clinical trials, median OS was 7.0 months for patients treated with immunotherapy and 7.9 months for patients treated with BRAF inhibitors. In "real world" studies, median OS was 4.3 months and 7.7 months for patients treated with immunotherapy and BRAF inhibitors, respectively. Evidence of clinical activity exists for both immunotherapy and MAP-kinase inhibitors. CONCLUSIONS MAP-kinase inhibitors and immunologic checkpoint blockade antibodies have clinical activity and may achieve improved OS in patients with metastatic melanoma and BM. These results support the inclusion of patients with BM in investigations of new agents and new treatment regimens for metastatic melanoma.
Collapse
Affiliation(s)
- Francesco Spagnolo
- Department of Plastic and Reconstructive Surgery, IRCCS AOU San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy.
| | - Virginia Picasso
- Department of Medical Oncology, U.O. Oncologia Medica 2, IRCCS AOU San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Matteo Lambertini
- Department of Medical Oncology, U.O. Oncologia Medica 2, IRCCS AOU San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Vincenzo Ottaviano
- Department of Plastic and Reconstructive Surgery, St George's Hospital, London, UK
| | - Beatrice Dozin
- Clinical Epidemiology Unit, IRCCS AOU San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Paola Queirolo
- Department of Medical Oncology, U.O. Oncologia Medica 2, IRCCS AOU San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| |
Collapse
|
5941
|
Saeed M, van Brakel M, Zalba S, Schooten E, Rens JAP, Koning GA, Debets R, Ten Hagen TLM. Targeting melanoma with immunoliposomes coupled to anti-MAGE A1 TCR-like single-chain antibody. Int J Nanomedicine 2016; 11:955-75. [PMID: 27022262 PMCID: PMC4792179 DOI: 10.2147/ijn.s96123] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Therapy of melanoma using T-cells with genetically introduced T-cell receptors (TCRs) directed against a tumor-selective cancer testis antigen (CTA) NY-ESO1 demonstrated clear antitumor responses in patients without side effects. Here, we exploited the concept of TCR-mediated targeting through introduction of single-chain variable fragment (scFv) antibodies that mimic TCRs in binding major histocompatibility complex-restricted CTA. We produced scFv antibodies directed against Melanoma AntiGEn A1 (MAGE A1) presented by human leukocyte antigen A1 (HLA-A1), in short M1/A1, and coupled these TCR-like antibodies to liposomes to achieve specific melanoma targeting. Two anti-M1/A1 antibodies with different ligand-binding affinities were derived from a phage-display library and reformatted into scFvs with an added cysteine at their carboxyl termini. Protein production conditions, ie, bacterial strain, temperature, time, and compartments, were optimized, and following production, scFv proteins were purified by immobilized metal ion affinity chromatography. Batches of pure scFvs were validated for specific binding to M1/A1-positive B-cells by flow cytometry. Coupling of scFvs to liposomes was conducted by employing different conditions, and an optimized procedure was achieved. In vitro experiments with immunoliposomes demonstrated binding of M1/A1-positive B-cells as well as M1/A1-positive melanoma cells and internalization by these cells using flow cytometry and confocal microscopy. Notably, the scFv with nonenhanced affinity of M1/A1, but not the one with enhanced affinity, was exclusively bound to and internalized by melanoma tumor cells expressing M1/A1. Taken together, antigen-mediated targeting of tumor cells as well as promoting internalization of nanoparticles by these tumor cells is mediated by TCR-like scFv and can contribute to melanoma-specific targeting.
Collapse
Affiliation(s)
- Mesha Saeed
- Laboratory of Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus MC, Rotterdam, the Netherlands
| | - Mandy van Brakel
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Sara Zalba
- Laboratory of Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus MC, Rotterdam, the Netherlands
| | - Erik Schooten
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Joost A P Rens
- Laboratory of Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus MC, Rotterdam, the Netherlands
| | - Gerben A Koning
- Laboratory of Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus MC, Rotterdam, the Netherlands
| | - Reno Debets
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Timo L M Ten Hagen
- Laboratory of Experimental Surgical Oncology, Section Surgical Oncology, Department of Surgery, Erasmus MC, Rotterdam, the Netherlands
| |
Collapse
|
5942
|
Strauss J, Madan RA. Therapeutic vaccines for prostate cancer: recent advances and future directions. Expert Rev Vaccines 2016; 15:907-14. [PMID: 26889831 DOI: 10.1586/14760584.2016.1155988] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In recent years, therapeutic cancer vaccines have emerged as a viable and promising treatment for prostate cancer. Beyond sipuleucel-T, phase III trials are evaluating multiple vaccine platforms in men with this disease. Growing data evaluating vaccine therapies suggests that these agents are more effective in patients with more indolent and possibly also earlier stages of disease. In addition, a wealth of preclinical data has shown that traditional prostate cancer treatments including anti androgens, cytotoxic and radiation therapies may provide immunologic synergy when given in combination with vaccine platforms. Building off this data, numerous clinical trials are evaluating therapeutic cancer vaccines in early stage prostate cancer and also in combination with traditional prostate cancer therapies. In addition, in order to optimize immune responses, ongoing trials are evaluating vaccines in combination with immune checkpoint inhibitors. Preliminary data from these trials have been promising and are offering an exciting glimpse at the future of immunotherapy for this disease.
Collapse
Affiliation(s)
- Julius Strauss
- a Genitourinary Malignancies Branch, Center for Cancer Research , National Cancer Institute , Bethesda , MD , USA
| | - Ravi A Madan
- a Genitourinary Malignancies Branch, Center for Cancer Research , National Cancer Institute , Bethesda , MD , USA
| |
Collapse
|
5943
|
Abdel-Rahman O, Fouad M. Risk of pneumonitis in cancer patients treated with immune checkpoint inhibitors: a meta-analysis. Ther Adv Respir Dis 2016; 10:183-93. [PMID: 26944362 DOI: 10.1177/1753465816636557] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND A meta-analysis of the risk of pneumonitis associated with the use of immune checkpoint inhibitors in cancer patients has been conducted. METHODS Eligible publications included randomized trials of cancer patients on immune checkpoint inhibitors, describing events of all-grade and high-grade pneumonitis. RESULTS After exclusion of noneligible citations, a total of 11 clinical trials were eligible for the meta-analysis. The odds ratio was 3.96 [95% confidence interval (CI): 2.02-7.79; p < 0.0001] for all-grade pneumonitis and 2.87 (95% CI: 0.90-9.20; p = 0.08) for high-grade pneumonitis. Moreover, the odds ratio of all-grade pneumonitis with a nivolumab/ipilimumab combination versus ipilimumab monotherapy was 3.68 (95% CI: 1.59-8.50; p = 0.002) and, for high-grade pneumonitis, it was 1.86(95% CI: 0.36-9.53; p = 0.46). Subgroup analysis did not reveal a difference between lung cancer patients and other cancer patients in the risk of pneumonitis. CONCLUSIONS Our analysis provided evidence that the use of immune checkpoint inhibitors is associated with an increased risk of all-grade pneumonitis compared with chemotherapy or placebo controls.
Collapse
Affiliation(s)
- Omar Abdel-Rahman
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Lotfy Elsayed Street, Cairo 11665, Egypt
| | - Mona Fouad
- Medical Microbiology and Immunology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
5944
|
Beers SA, Glennie MJ, White AL. Influence of immunoglobulin isotype on therapeutic antibody function. Blood 2016; 127:1097-101. [PMID: 26764357 PMCID: PMC4797141 DOI: 10.1182/blood-2015-09-625343] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/09/2016] [Indexed: 12/28/2022] Open
Abstract
Monoclonal antibody (mAb) therapeutics are revolutionizing cancer treatment; however, not all tumors respond, and agent optimization is essential to improve outcome. It has become clear over recent years that isotype choice is vital to therapeutic success with agents that work through different mechanisms, direct tumor targeting, agonistic receptor engagement, or receptor-ligand blockade, having contrasting requirements. Here we summarize how isotype dictates mAb activity and discuss ways in which this information can be used for the development of enhanced therapeutics.
Collapse
Affiliation(s)
- Stephen A Beers
- Antibody and Vaccine Group, Cancer Sciences Unit, University of Southampton, Faculty of Medicine, General Hospital, Southampton, United Kingdom
| | - Martin J Glennie
- Antibody and Vaccine Group, Cancer Sciences Unit, University of Southampton, Faculty of Medicine, General Hospital, Southampton, United Kingdom
| | - Ann L White
- Antibody and Vaccine Group, Cancer Sciences Unit, University of Southampton, Faculty of Medicine, General Hospital, Southampton, United Kingdom
| |
Collapse
|
5945
|
Zou W, Wolchok JD, Chen L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci Transl Med 2016; 8:328rv4. [PMID: 26936508 PMCID: PMC4859220 DOI: 10.1126/scitranslmed.aad7118] [Citation(s) in RCA: 1859] [Impact Index Per Article: 206.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PD-L1 and PD-1 (PD) pathway blockade is a highly promising therapy and has elicited durable antitumor responses and long-term remissions in a subset of patients with a broad spectrum of cancers. How to improve, widen, and predict the clinical response to anti-PD therapy is a central theme in the field of cancer immunology and immunotherapy. Oncologic, immunologic, genetic, and biological studies focused on the human cancer microenvironment have yielded substantial insight into this issue. Here, we focus on tumor microenvironment and evaluate several potential therapeutic response markers including the PD-L1 and PD-1 expression pattern, genetic mutations within cancer cells and neoantigens, cancer epigenetics and effector T cell landscape, and microbiota. We further clarify the mechanisms of action of these markers and their roles in shaping, being shaped, and/or predicting therapeutic responses. We also discuss a variety of combinations with PD pathway blockade and their scientific rationales for cancer treatment.
Collapse
Affiliation(s)
- Weiping Zou
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA.
| | - Jedd D Wolchok
- Department of Medicine and the Ludwig Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Lieping Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven CT 06519, USA.
| |
Collapse
|
5946
|
Ludwig ML, Birkeland AC, Hoesli R, Swiecicki P, Spector ME, Brenner JC. Changing the paradigm: the potential for targeted therapy in laryngeal squamous cell carcinoma. Cancer Biol Med 2016; 13:87-100. [PMID: 27144065 PMCID: PMC4850131 DOI: 10.28092/j.issn.2095-3941.2016.0010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 02/17/2016] [Indexed: 01/05/2023] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) remains a highly morbid and fatal disease. Historically, it has been a model example for organ preservation and treatment stratification paradigms. Unfortunately, survival for LSCC has stagnated over the past few decades. As the era of next-generation sequencing and personalized treatment for cancer approaches, LSCC may be an ideal disease for consideration of further treatment stratification and personalization. Here, we will discuss the important history of LSCC as a model system for organ preservation, unique and potentially targetable genetic signatures of LSCC, and methods for bringing stratified, personalized treatment strategies to the 21(st) century.
Collapse
Affiliation(s)
- Megan L. Ludwig
- Department of Otolaryngology, Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Andrew C. Birkeland
- Department of Otolaryngology, Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Rebecca Hoesli
- Department of Otolaryngology, Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Paul Swiecicki
- Department of Hematology Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Matthew E. Spector
- Department of Otolaryngology, Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - J. Chad Brenner
- Department of Otolaryngology, Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
5947
|
Garon EB. The race for combined checkpoint inhibition in NSCLC. Lancet Oncol 2016; 17:259-260. [DOI: 10.1016/s1470-2045(15)00580-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 12/03/2015] [Indexed: 10/22/2022]
|
5948
|
White ML, Atwell TD, Kurup AN, Schmit GD, Carter RE, Geske JR, Kottschade LA, Pulido JS, Block MS, Jakub JW, Callstrom MR, Markovic SN. Recurrence and Survival Outcomes After Percutaneous Thermal Ablation of Oligometastatic Melanoma. Mayo Clin Proc 2016; 91:288-96. [PMID: 26827235 DOI: 10.1016/j.mayocp.2015.10.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 09/15/2015] [Accepted: 10/23/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVES To evaluate focal treatment of melanoma metastases and to explore whether any potential extended survival benefit exists in a select patient population. PATIENTS AND METHODS All patients who underwent image-guided local thermal ablation of metastatic melanoma over an 11-year period (January 1, 2002, to December 31, 2013) were retrospectively identified using an internally maintained clinical registry. Only patients with oligometastatic stage IV disease amenable to complete ablation of all clinical disease at the time of ablation were included in the analysis. Overall survival and median progression-free survival periods were calculated. RESULTS Thirty-three patients with primary ocular or nonocular melanoma had 66 metastases treated in the lungs, liver, bones, or soft tissues. Eleven (33%) patients were on systemic medical therapy at the time of the procedure. The median survival time was 3.8 years (range, 0.5-10.5 years), with a 4-year estimated survival of 44.1% (95% CI, 28%-68%). Local recurrence at the ablation site developed in 15.1% (5 of 33) of the patients and 13.6% of the tumors (9 of 66). The median progression-free survival time was 4.4 months (95% CI, 1.4 months to 10.5 years), with an estimated 1-year progression-free survival of 30.3% (95% CI, 18%-51%). A subgroup analysis identified 11 patients with primary ocular melanoma and 22 with nonocular melanoma, with a median survival time of 3.9 years (range, 0.9-4.7 years) and 3.8 years (range, 0.5-10.5 years), respectively (P=.58). There were no major complications and no deaths within 30 days of the procedure. CONCLUSION Selective use of image-guided thermal ablation of oligometastatic melanoma may provide results similar to surgical resection in terms of technical effectiveness and oncologic outcomes with minimal risk.
Collapse
Affiliation(s)
| | | | | | | | - Rickey E Carter
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN
| | - Jennifer R Geske
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN
| | | | - Jose S Pulido
- Department of Opthalmology, Mayo Clinic, Rochester, MN
| | | | - James W Jakub
- Division of Subspecialty General Surgery, Mayo Clinic, Rochester, MN
| | | | | |
Collapse
|
5949
|
Deel A. Nivolumab in Metastatic Non-Small Cell Lung Cancer. J Adv Pract Oncol 2016; 7:220-225. [PMID: 28090370 PMCID: PMC5226313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
5950
|
Girotti MR, Gremel G, Lee R, Galvani E, Rothwell D, Viros A, Mandal AK, Lim KHJ, Saturno G, Furney SJ, Baenke F, Pedersen M, Rogan J, Swan J, Smith M, Fusi A, Oudit D, Dhomen N, Brady G, Lorigan P, Dive C, Marais R. Application of Sequencing, Liquid Biopsies, and Patient-Derived Xenografts for Personalized Medicine in Melanoma. Cancer Discov 2016; 6:286-99. [PMID: 26715644 DOI: 10.1158/2159-8290.cd-15-1336] [Citation(s) in RCA: 183] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/23/2015] [Indexed: 11/16/2022]
Abstract
UNLABELLED Targeted therapies and immunotherapies have transformed melanoma care, extending median survival from ∼9 to over 25 months, but nevertheless most patients still die of their disease. The aim of precision medicine is to tailor care for individual patients and improve outcomes. To this end, we developed protocols to facilitate individualized treatment decisions for patients with advanced melanoma, analyzing 364 samples from 214 patients. Whole exome sequencing (WES) and targeted sequencing of circulating tumor DNA (ctDNA) allowed us to monitor responses to therapy and to identify and then follow mechanisms of resistance. WES of tumors revealed potential hypothesis-driven therapeutic strategies for BRAF wild-type and inhibitor-resistant BRAF-mutant tumors, which were then validated in patient-derived xenografts (PDX). We also developed circulating tumor cell-derived xenografts (CDX) as an alternative to PDXs when tumors were inaccessible or difficult to biopsy. Thus, we describe a powerful technology platform for precision medicine in patients with melanoma. SIGNIFICANCE Although recent developments have revolutionized melanoma care, most patients still die of their disease. To improve melanoma outcomes further, we developed a powerful precision medicine platform to monitor patient responses and to identify and validate hypothesis-driven therapies for patients who do not respond, or who develop resistance to current treatments.
Collapse
Affiliation(s)
- Maria Romina Girotti
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Gabriela Gremel
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Rebecca Lee
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Elena Galvani
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Dominic Rothwell
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Amaya Viros
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Amit Kumar Mandal
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Kok Haw Jonathan Lim
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Grazia Saturno
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Simon J Furney
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Franziska Baenke
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Malin Pedersen
- Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| | - Jane Rogan
- The University of Manchester, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Jacqueline Swan
- Research Services, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Matthew Smith
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Alberto Fusi
- The University of Manchester, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Deemesh Oudit
- The University of Manchester, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Nathalie Dhomen
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Ged Brady
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Paul Lorigan
- The University of Manchester, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Caroline Dive
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom
| | - Richard Marais
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester, United Kingdom.
| |
Collapse
|