101
|
Resistance to combination BRAF and MEK inhibition in metastatic melanoma: Where to next? Eur J Cancer 2016; 62:76-85. [DOI: 10.1016/j.ejca.2016.04.005] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 04/05/2016] [Indexed: 12/12/2022]
|
102
|
Moving Immune Checkpoint Blockade in Thoracic Tumors beyond NSCLC. J Thorac Oncol 2016; 11:1819-1836. [PMID: 27288978 DOI: 10.1016/j.jtho.2016.05.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 05/24/2016] [Accepted: 05/28/2016] [Indexed: 02/07/2023]
Abstract
SCLC and malignant pleural mesothelioma (MPM) are historically characterized by a disappointing lack of significant therapeutic breakthroughs for novel agents, and both malignancies represent true unmet medical needs. Given the promising results of anti-cytotoxic T-lymphocyte associated protein-4 and anti-programmed cell death-1/programmed death ligand-1 antibodies in the treatment of advanced NSCLCs, these immune checkpoint inhibitors are now also under investigation in SCLC and MPM, as well as in thymic epithelial tumors (TETs). Here, we review the biological and clinical rationale for immune checkpoint inhibition in SCLC, MPM, and TETs and present preliminary clinical results with available antibodies. Immunotherapeutic perspectives for these malignancies in terms of novel agents currently under evaluation or anticipated in the near future are also discussed. Current immune checkpoint blockers targeting cytotoxic T-lymphocyte associated protein-4 and the programmed cell death-1/programmed death ligand-1 axis, administered alone or in combination and as multimodality treatment, are likely to be a valuable addition to the therapeutic array for managing SCLC and MPM; studies in TETs, which are currently in their infancy, are merited. Close attention to potential toxicities will be important to the success of such strategies in these settings.
Collapse
|
103
|
Targeted Therapy and Checkpoint Immunotherapy Combinations for the Treatment of Cancer. Trends Immunol 2016; 37:462-476. [PMID: 27216414 DOI: 10.1016/j.it.2016.04.010] [Citation(s) in RCA: 204] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 04/27/2016] [Accepted: 04/29/2016] [Indexed: 12/11/2022]
Abstract
Many advances in the treatment of cancer have been driven by the development of targeted therapies that inhibit oncogenic signaling pathways and tumor-associated angiogenesis, as well as by the recent development of therapies that activate a patient's immune system to unleash antitumor immunity. Some targeted therapies can have effects on host immune responses, in addition to their effects on tumor biology. These immune-modulating effects, such as increasing tumor antigenicity or promoting intratumoral T cell infiltration, provide a rationale for combining these targeted therapies with immunotherapies. Here, we discuss the immune-modulating effects of targeted therapies against the MAPK and VEGF signaling pathways, and how they may synergize with immunomodulatory antibodies that target PD1/PDL1 and CTLA4. We critically examine the rationale in support of these combinations in light of the current understanding of the underlying mechanisms of action of these therapies. We also discuss the available preclinical and clinical data for these combination approaches and their implications regarding mechanisms of action. Insights from these studies provide a framework for considering additional combinations of targeted therapies and immunotherapies for the treatment of cancer.
Collapse
|
104
|
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: 384] [Impact Index Per Article: 48.0] [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
|
105
|
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
|
106
|
Expression of programmed cell death ligand 1 (PD-L1) and prevalence of tumor-infiltrating lymphocytes (TILs) in chordoma. Oncotarget 2016; 6:11139-49. [PMID: 25871477 PMCID: PMC4484445 DOI: 10.18632/oncotarget.3576] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 02/21/2015] [Indexed: 12/31/2022] Open
Abstract
Chordomas are primary malignant tumors of the notochord that are resistant to conventional chemotherapy. Expression of programmed cell death ligand 1 (PD-L1), prevalence of tumor-infiltrating lymphocytes (TILs), and their clinical relevance in chordoma remain unknown. We evaluated PD-L1 expression in three chordoma cell lines and nine chordoma tissue samples by western blot. Immunohistochemical staining was performed on a chordoma tissue microarray (TMA) that contained 78 tissue specimens. We also correlated the expression of PD-L1 and TILs with clinical outcomes. PD-L1 protein expression was demonstrated to be induced by IFN-γ in both UCH1 and UCH2 cell lines. Across nine human chordoma tissue samples, PD-L1 protein was differentially expressed. 94.9% of chordoma samples showed positive PD-L1 expression in the TMA. The expression score of PD-L1 for metastatic chordoma tumors was significant higher as compared with non-metastatic chordoma tumors. Expression of PD-L1 protein significantly correlates with the presence of elevated TILs, which correlates with metastasis. In summary, our study showed high levels of PD-L1 are expressed in chordoma, which is correlated with the prevalence of TILs. The current study suggests targeting PD-L1 may be a novel immunotherapeutic strategy for chordoma clinical trials.
Collapse
|
107
|
Kong BY, Carlino MS, Menzies AM. Biology and treatment of BRAF mutant metastatic melanoma. Melanoma Manag 2016; 3:33-45. [PMID: 30190871 DOI: 10.2217/mmt.15.38] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 10/12/2015] [Indexed: 12/31/2022] Open
Abstract
BRAF inhibitors were among the first systemic therapies to show clinical benefit in metastatic melanoma. Here, we review the spectrum of BRAF mutations in melanoma, their role in oncogenesis, clinicopathological associations and response to treatment. The differing biology and clinical features of V600E- and V600K-mutated melanoma are outlined. The molecular changes associated with BRAF fusion genes and their response to targeted therapies, as well as the role of immunotherapy in treatment sequencing with targeted therapies are discussed.
Collapse
Affiliation(s)
- Benjamin Y Kong
- Crown Princess Mary Cancer Care Centre, Westmead, Sydney, NSW 2145, Australia.,Crown Princess Mary Cancer Care Centre, Westmead, Sydney, NSW 2145, Australia
| | - Matteo S Carlino
- Crown Princess Mary Cancer Care Centre, Westmead, Sydney, NSW 2145, Australia.,Melanoma Institute Australia, North Sydney, NSW 2060, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia.,Crown Princess Mary Cancer Care Centre, Westmead, Sydney, NSW 2145, Australia.,Melanoma Institute Australia, North Sydney, NSW 2060, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, North Sydney, NSW 2060, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia.,Royal North Shore Hospital, Sydney, NSW 2065, Australia.,Melanoma Institute Australia, North Sydney, NSW 2060, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia.,Royal North Shore Hospital, Sydney, NSW 2065, Australia
| |
Collapse
|
108
|
Cooper ZA, Reuben A, Spencer CN, Prieto PA, Austin-Breneman JL, Jiang H, Haymaker C, Gopalakrishnan V, Tetzlaff MT, Frederick DT, Sullivan RJ, Amaria RN, Patel SP, Hwu P, Woodman SE, Glitza IC, Diab A, Vence LM, Rodriguez-Canales J, Parra ER, Wistuba II, Coussens LM, Sharpe AH, Flaherty KT, Gershenwald JE, Chin L, Davies MA, Clise-Dwyer K, Allison JP, Sharma P, Wargo JA. Distinct clinical patterns and immune infiltrates are observed at time of progression on targeted therapy versus immune checkpoint blockade for melanoma. Oncoimmunology 2016; 5:e1136044. [PMID: 27141370 PMCID: PMC4839346 DOI: 10.1080/2162402x.2015.1136044] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 12/17/2015] [Accepted: 12/18/2015] [Indexed: 12/20/2022] Open
Abstract
We have made major advances in the treatment of melanoma through the use of targeted therapy and immune checkpoint blockade; however, clinicians are posed with therapeutic dilemmas regarding timing and sequence of therapy. There is a growing appreciation of the impact of antitumor immune responses to these therapies, and we performed studies to test the hypothesis that clinical patterns and immune infiltrates differ at progression on these treatments. We observed rapid clinical progression kinetics in patients on targeted therapy compared to immune checkpoint blockade. To gain insight into possible immune mechanisms behind these differences, we performed deep immune profiling in tumors of patients on therapy. We demonstrated low CD8+ T-cell infiltrate on targeted therapy and high CD8+ T-cell infiltrate on immune checkpoint blockade at clinical progression. These data have important implications, and suggest that antitumor immune responses should be assessed when considering therapeutic options for patients with melanoma.
Collapse
Affiliation(s)
- Zachary A Cooper
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexandre Reuben
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Christine N Spencer
- Genomic Medicine, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Peter A Prieto
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Jacob L Austin-Breneman
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Hong Jiang
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Cara Haymaker
- Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | | | - Michael T Tetzlaff
- Pathology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Dennie T Frederick
- Division of Medical Oncology, Massachusetts General Hospital , Boston, MA, USA
| | - Ryan J Sullivan
- Division of Medical Oncology, Massachusetts General Hospital , Boston, MA, USA
| | - Rodabe N Amaria
- Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Sapna P Patel
- Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Patrick Hwu
- Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Scott E Woodman
- Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Isabella C Glitza
- Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Adi Diab
- Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Luis M Vence
- Immunology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Jaime Rodriguez-Canales
- Translational Molecular Pathology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Edwin R Parra
- Translational Molecular Pathology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Ignacio I Wistuba
- Translational Molecular Pathology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Lisa M Coussens
- Department of Cell, Developmental and Cancer Biology and Knight Cancer Institute, Oregon Health and Science University , Portland, OR, USA
| | - Arlene H Sharpe
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School , Boston, MA, USA
| | - Keith T Flaherty
- Division of Medical Oncology, Massachusetts General Hospital , Boston, MA, USA
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Lynda Chin
- Genomic Medicine, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Michael A Davies
- Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Karen Clise-Dwyer
- Stem Cell Transplantation, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - James P Allison
- Immunology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Padmanee Sharma
- Immunology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer A Wargo
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
109
|
Kakavand H, Wilmott JS, Long GV, Scolyer RA. Targeted therapies and immune checkpoint inhibitors in the treatment of metastatic melanoma patients: a guide and update for pathologists. Pathology 2016; 48:194-202. [PMID: 27020392 DOI: 10.1016/j.pathol.2015.12.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 11/28/2015] [Indexed: 12/19/2022]
Abstract
The previously dismal prospects for patients with advanced stage metastatic melanoma have greatly improved in recent years. Enhanced understanding of both the pathogenesis of melanoma and its molecular drivers, as well as the importance and regulation of anti-tumour immune responses, have provided new therapeutic opportunities for melanoma patients. There are two major distinct categories of systemic treatments with activity for patients with metastatic melanoma: (1) targeted therapies, which act to inhibit the oncogenes that drive the aberrant growth and dissemination of the tumour; and (2) immune checkpoint inhibitor therapies, which act to enhance anti-tumour immune responses by blocking negative regulators of immunity. Pathologists play a critical and expanding role in the selection of the most appropriate treatment for individual metastatic melanoma patients in the modern era of personalised/precision medicine. The molecular pathology testing of melanoma tumour tissue for the presence of targetable oncogenic mutations is already part of routine practice in many institutions. In addition, other potential oncogenic therapeutic targets continue to be identified and pathology testing techniques must readily adapt to this rapidly changing field. Recent research findings suggest that pathological assessment of tumour associated immune cells and immunosuppressive ligand expression of the tumour are likely to be important in identifying patients most likely to benefit from immune checkpoint inhibitors. Similarly, pathological and molecular observations of on-treatment tumour tissue biopsies taken from patients on targeted therapies have provided new insights into the mechanisms of action of targeted molecular therapies, have contributed to the identification of resistance mechanisms to these novel therapies and may be of higher value for selecting patients most likely to benefit from therapies. These data have already provided a rational biological basis for the exciting prospect of combining them to further improve survival rates and this is currently being investigated in clinical trials. Ultimately it may be the responsibility of the pathologist to identify which therapy or combination of therapies is most likely to benefit individual patients.
Collapse
Affiliation(s)
- Hojabr Kakavand
- Melanoma Institute Australia, North Sydney, Australia; The University of Sydney, Sydney, Australia
| | - James S Wilmott
- Melanoma Institute Australia, North Sydney, Australia; The University of Sydney, Sydney, Australia
| | - Georgina V Long
- Melanoma Institute Australia, North Sydney, Australia; The University of Sydney, Sydney, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, North Sydney, Australia; The University of Sydney, Sydney, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia.
| |
Collapse
|
110
|
Potential biomarker for checkpoint blockade immunotherapy and treatment strategy. Tumour Biol 2016; 37:4251-61. [PMID: 26779629 DOI: 10.1007/s13277-016-4812-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/06/2016] [Indexed: 12/12/2022] Open
Abstract
Programmed cell death protein-1 (PD-1) and ligand (PD-L1) provide an important escape mechanism from immune attack, and blockade therapy of these proteins show promising clinical benefits in many types of cancer. PD-L1 can be induced by interferon-gamma (IFN-γ), hypoxia, or toll-like receptor (TLR)-mediated pathways that confer adaptive immune resistance, or upregulated by oncogenic signals leading to constitutive expression and resulting in intrinsic immune resistance. The PD-1/PD-L1 checkpoint blockade, which targets regulatory pathways in T cells to overcome immune resistance, is correlated to PD-L1 expression pattern and the presence of tumor-infiltrating lymphocytes (TILs). Meanwhile, immunogenic mutation loads show significant response to checkpoint blockade, which is probably due to PD-1/L1 status and TIL content. Finally, the clinical strategies to design effective checkpoint-targeting immunotherapies are based on the classification of inducible/constitutive expression of PD-L1 and the presence of TILs.
Collapse
|
111
|
Galluzzi L, Buqué A, Kepp O, Zitvogel L, Kroemer G. Immunological Effects of Conventional Chemotherapy and Targeted Anticancer Agents. Cancer Cell 2015; 28:690-714. [PMID: 26678337 DOI: 10.1016/j.ccell.2015.10.012] [Citation(s) in RCA: 1112] [Impact Index Per Article: 123.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 10/05/2015] [Accepted: 10/23/2015] [Indexed: 11/23/2022]
Abstract
The tremendous clinical success of checkpoint blockers illustrates the potential of reestablishing latent immunosurveillance for cancer therapy. Although largely neglected in the clinical practice, accumulating evidence indicates that the efficacy of conventional and targeted anticancer agents does not only involve direct cytostatic/cytotoxic effects, but also relies on the (re)activation of tumor-targeting immune responses. Chemotherapy can promote such responses by increasing the immunogenicity of malignant cells, or by inhibiting immunosuppressive circuitries that are established by developing neoplasms. These immunological "side" effects of chemotherapy are desirable, and their in-depth comprehension will facilitate the design of novel combinatorial regimens with improved clinical efficacy.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Equipe 11 Labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France; INSERM, U1138, 75006 Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France; Université Pierre et Marie Curie/Paris VI, 75006 Paris, France; Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France
| | - Aitziber Buqué
- Equipe 11 Labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France; INSERM, U1138, 75006 Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France; Université Pierre et Marie Curie/Paris VI, 75006 Paris, France; Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France
| | - Oliver Kepp
- Equipe 11 Labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France; INSERM, U1138, 75006 Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France; Université Pierre et Marie Curie/Paris VI, 75006 Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France; INSERM, U1015, 94805 Villejuif, France; Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 507, 94805 Villejuif, France; Université Paris Sud/Paris XI, 94270 Le Kremlin-Bicêtre, France.
| | - Guido Kroemer
- Equipe 11 Labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France; INSERM, U1138, 75006 Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France; Université Pierre et Marie Curie/Paris VI, 75006 Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France; Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, 17176 Stockholm, Sweden.
| |
Collapse
|
112
|
Johnson DB, Menzies AM, Zimmer L, Eroglu Z, Ye F, Zhao S, Rizos H, Sucker A, Scolyer RA, Gutzmer R, Gogas H, Kefford RF, Thompson JF, Becker JC, Berking C, Egberts F, Loquai C, Goldinger SM, Pupo GM, Hugo W, Kong X, Garraway LA, Sosman JA, Ribas A, Lo RS, Long GV, Schadendorf D. Acquired BRAF inhibitor resistance: A multicenter meta-analysis of the spectrum and frequencies, clinical behaviour, and phenotypic associations of resistance mechanisms. Eur J Cancer 2015; 51:2792-9. [PMID: 26608120 PMCID: PMC4666799 DOI: 10.1016/j.ejca.2015.08.022] [Citation(s) in RCA: 242] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 08/22/2015] [Indexed: 11/15/2022]
Abstract
BACKGROUND Acquired resistance to BRAF inhibitors (BRAFi) is a near-universal phenomenon caused by numerous genetic and non-genetic alterations. In this study, we evaluated the spectrum, onset, pattern of progression, and subsequent clinical outcomes associated with specific mechanisms of resistance. METHODS We compiled clinical and genetic data from 100 patients with 132 tissue samples obtained at progression on BRAFi therapy from 3 large, previously published studies of BRAFi resistance. These samples were subjected to whole-exome sequencing and/or polymerase chain reaction-based genetic testing. RESULTS Among 132 samples, putative resistance mechanisms were identified in 58%, including NRAS or KRAS mutations (20%), BRAF splice variants (16%), BRAF(V600E/K) amplifications (13%), MEK1/2 mutations (7%), and non-mitogen-activated protein kinase pathway alterations (11%). Marked heterogeneity was observed within tumors and patients; 18 of 19 patients (95%) with more than one progression biopsy had distinct/unknown drivers of resistance between samples. NRAS mutations were associated with vemurafenib use (p = 0.045) and intracranial metastases (p = 0.036), and MEK1/2 mutations correlated with hepatic progression (p = 0.011). Progression-free survival and overall survival were similar across resistance mechanisms. The median survival after disease progression was 6.9 months, and responses to subsequent BRAF and MEK inhibition were uncommon (2 of 15; 13%). Post-progression outcomes did not correlate with specific acquired BRAFi-resistance mechanisms. CONCLUSIONS This is the first study to systematically characterise the clinical implications of particular acquired BRAFi-resistance mechanisms in patients with BRAF-mutant melanoma largest study to compile the landscape of resistance. Despite marked heterogeneity of resistance mechanisms within patients, NRAS mutations correlated with vemurafenib use and intracranial disease involvement.
Collapse
Affiliation(s)
- Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Alexander M Menzies
- Melanoma Institute Australia, Sydney, NSW, Australia; Centre for Cancer Research, The University of Sydney at The Westmead Millennium Institute, Westmead, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Lisa Zimmer
- Department of Dermatology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Zeynep Eroglu
- Department of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA; Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Fei Ye
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shilin Zhao
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Helen Rizos
- Centre for Cancer Research, The University of Sydney at The Westmead Millennium Institute, Westmead, NSW, Australia; Macquarie University, Sydney, NSW, Australia
| | - Antje Sucker
- Department of Dermatology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Richard A Scolyer
- Melanoma Institute Australia, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Ralf Gutzmer
- Department of Dermatology and Allergy, Skin Cancer Center Hannover, Hannover Medical School, Hannover, Germany
| | - Helen Gogas
- First Department of Medicine, University of Athens Medical School, Athens, Greece
| | - Richard F Kefford
- Melanoma Institute Australia, Sydney, NSW, Australia; Centre for Cancer Research, The University of Sydney at The Westmead Millennium Institute, Westmead, NSW, Australia; Macquarie University, Sydney, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - John F Thompson
- Melanoma Institute Australia, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Jürgen C Becker
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| | - Carola Berking
- Department of Dermatology, Ludwig-Maximilians-University, Munich, Germany
| | - Friederike Egberts
- Department of Dermatology, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Carmen Loquai
- Department of Dermatology, University Medical Center, Mainz, Germany
| | | | - Gulietta M Pupo
- Centre for Cancer Research, The University of Sydney at The Westmead Millennium Institute, Westmead, NSW, Australia
| | - Willy Hugo
- Department of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Xiangju Kong
- Department of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Levi A Garraway
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jeffrey A Sosman
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Antoni Ribas
- Department of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Roger S Lo
- Department of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Georgina V Long
- Melanoma Institute Australia, Sydney, NSW, Australia; Centre for Cancer Research, The University of Sydney at The Westmead Millennium Institute, Westmead, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
113
|
Kakavand H, Vilain RE, Wilmott JS, Burke H, Yearley JH, Thompson JF, Hersey P, Long GV, Scolyer RA. Tumor PD-L1 expression, immune cell correlates and PD-1+ lymphocytes in sentinel lymph node melanoma metastases. Mod Pathol 2015; 28:1535-44. [PMID: 26403784 DOI: 10.1038/modpathol.2015.110] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Revised: 08/11/2015] [Accepted: 08/29/2015] [Indexed: 12/22/2022]
Abstract
Melanoma patients with sentinel lymph node metastases have variable 5-year survival rates (39-70%). The prognostic significance of tumor-infiltrating lymphocytes in sentinel lymph node metastases from such patients is currently unknown. Anti-PD-1/PD-L1 inhibitors have significantly improved clinical outcome in unresectable AJCC stage IIIC/IV metastatic melanoma patients, and are being trialed in the adjuvant setting in advanced stage disease, however, their role in early stage (sentinel lymph node positive) metastatic disease remains unclear. The aims of this study were to characterize, in sentinel lymph nodes, the subpopulations of lymphocytes that interact with metastatic melanoma cells and analyze their associations with outcome, and to determine tumor PD-L1 expression as this may provide a rational scientific basis for the administration of adjuvant anti-PD-1/PD-L1 inhibitors in sentinel lymph node positive metastatic melanoma patients. Sentinel lymph nodes containing metastatic melanoma from 60 treatment-naive patients were analyzed for CD3, CD4, CD8, FOXP3, PD-1, and PD-L1 using immunohistochemistry on serial sections. The results were correlated with clinicopathologic features and outcome. Positive correlations between recurrence-free/overall survival with the number of CD3+ tumor-infiltrating lymphocytes (hazard ratio=0.36 (0.17-0.76), P=0.005; hazard ratio=0.29 (0.14-0.61), P=0.0005, respectively), the number of CD4+ tumor-infiltrating lymphocytes (hazard ratio=0.34 (0.15-0.77), P=0.007; hazard ratio=0.32 (0.14-0.74), P=0.005, respectively), and the number of CD8+ tumor-infiltrating lymphocytes (hazard ratio =0.42 (0.21-0.85), P=0.013; hazard ratio =0.32 (0.19-0.78), P=0.006, respectively) were observed. There was also a negative correlation with the number of peritumoral PD-1+ lymphocytes (hazard ratio=2.67 (1.17-6.13), P=0.016; hazard ratio=2.74 (1.14-6.76), P=0.019, respectively). Tumoral PD-L1 expression was present in 26 cases (43%) but did not correlate with outcome. The findings suggest that T-cell subsets in sentinel lymph node metastases can predict melanoma patient outcome. In addition, the relatively high number of PD-L1 positive sentinel lymph node melanoma metastases provides a rationale for anti-PD-1 therapy trials in sentinel lymph node positive melanoma patients, particularly those with peritumoral PD-1+ lymphocytes.
Collapse
Affiliation(s)
- Hojabr Kakavand
- Melanoma Institute Australia, North Sydney, NSW, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Ricardo E Vilain
- Melanoma Institute Australia, North Sydney, NSW, Australia.,Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - James S Wilmott
- Melanoma Institute Australia, North Sydney, NSW, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Hazel Burke
- Melanoma Institute Australia, North Sydney, NSW, Australia
| | | | - John F Thompson
- Melanoma Institute Australia, North Sydney, NSW, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Peter Hersey
- Melanoma Institute Australia, North Sydney, NSW, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Royal North Shore Hospital, St. Leonards, NSW, Australia
| | - Georgina V Long
- Melanoma Institute Australia, North Sydney, NSW, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, North Sydney, NSW, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.,Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| |
Collapse
|
114
|
Kim HS, Lee JY, Lim SH, Park K, Sun JM, Ko YH, Baek CH, Son YI, Jeong HS, Ahn YC, Lee MY, Hong M, Ahn MJ. Association Between PD-L1 and HPV Status and the Prognostic Value of PD-L1 in Oropharyngeal Squamous Cell Carcinoma. Cancer Res Treat 2015; 48:527-36. [PMID: 26511814 PMCID: PMC4843713 DOI: 10.4143/crt.2015.249] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 08/13/2015] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Oropharyngeal squamous cell carcinoma (OSCC) has been recognized as an immunosuppressive disease. Various mechanisms have been proposed for immune escape, including dysregulation of immune checkpoints such as the PD-1:PD-L1 pathway. We investigated the expression of programmed cell death-ligand 1 (PD-L1) in HPV-negative and HPV-positive OSCC to determine its prevalence and prognostic relevance. MATERIALS AND METHODS Using immunohistochemistry, 133 cases of OSCC were evaluated for expression of PD-L1. Formalin-fixed paraffin-embedded tumor samples were stained with monoclonal antibody (clone 5H1) to PD-L1. PD-L1 positivity was defined as membrane staining in ≥20% of tumor cells. Correlations between PD-L1 expression and HPV status and survival parameters were analyzed. RESULTS Of the 133 patients, 68% showed PD-L1 expression, and 67% of patients were positive for p16 expression by immunohistochemistry. No significant difference in PD-L1 expression was observed between HPV(-) and HPV(+) tumors (61% vs. 71%, p=0.274). No significant difference in age, gender, smoking history, location of tumor origin, or stage was observed according to PD-L1 status. With a median follow-up period of 44 months, older age (≥65) (p=0.017) and T3-4 stage (p<0.001) were associated with poor overall survival (OS), whereas PD-L1 expression did not affect OS in univariate and multivariate analysis. CONCLUSION PD-L1 expression was observed in the majority of OSCC patients regardless of HPV status. Further large prospective studies are required to determine the role of PD-L1 expression as a prognostic or predictive biomarker, and clinical studies of immune checkpoint inhibitors in OCSS are warranted regardless of HPV status.
Collapse
Affiliation(s)
- Hae Su Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Yun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sung Hee Lim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Keunchil Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jong-Mu Sun
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young Hyeh Ko
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Chung-Hwan Baek
- Department of Otorhinolaryngology-Head and Neck Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young-ik Son
- Department of Otorhinolaryngology-Head and Neck Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Han Sin Jeong
- Department of Otorhinolaryngology-Head and Neck Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yong Chan Ahn
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Min-Young Lee
- Division of Hematology-Oncology, Department of Internal Medicine, Soonchunhyang University Hospital, Soonchunhyang University College of Medicine, Seoul, Korea
| | - Mineui Hong
- Department of Pathology, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
115
|
Chinai JM, Janakiram M, Chen F, Chen W, Kaplan M, Zang X. New immunotherapies targeting the PD-1 pathway. Trends Pharmacol Sci 2015; 36:587-95. [PMID: 26162965 DOI: 10.1016/j.tips.2015.06.005] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/15/2015] [Accepted: 06/17/2015] [Indexed: 12/22/2022]
Abstract
Ligands from the B7 family bind to receptors of the CD28 family, which regulate early T cell activation in lymphoid organs and control inflammation and autoimmunity in peripheral tissues. Programmed death-1 (PD-1), a member of the CD28 family, is an inhibitory receptor on T cells and is responsible for their dysfunction in infectious diseases and cancers. The complex mechanisms controlling the expression and signaling of PD-1 and programmed death ligand 1 (PD-L1) are emerging. Recently completed and ongoing clinical trials that target these molecules have shown remarkable success by generating durable clinical responses in some cancer patients. In chronic viral infections, preclinical data reveal that targeting PD-1 and its ligands can improve T cell responses and virus clearance. There is also promise in stimulating this pathway for the treatment of autoimmune and inflammatory disorders.
Collapse
Affiliation(s)
- Jordan M Chinai
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Murali Janakiram
- Department of Oncology, Montefiore Medical Center, New York, NY 10467, USA
| | - Fuxiang Chen
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Wantao Chen
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Mark Kaplan
- Centers for Therapeutic Innovation, Pfizer Inc., New York, NY 10016, USA
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY 10461, USA; Department of Oncology, Montefiore Medical Center, New York, NY 10467, USA.
| |
Collapse
|
116
|
Cooper ZA, Reuben A, Austin-Breneman J, Wargo JA. Does It MEK a Difference? Understanding Immune Effects of Targeted Therapy. Clin Cancer Res 2015; 21:3102-4. [PMID: 26025561 DOI: 10.1158/1078-0432.ccr-15-0363] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 03/10/2015] [Indexed: 11/16/2022]
Abstract
BRAF inhibitor (BRAFi) treatment enhances antitumor immunity, but is associated with increased intratumoral PD-L1 expression. MEK inhibitors (MEKi) may alter T-cell function; however, recent studies demonstrate preserved T-cell infiltrate during treatment with BRAFi/MEKi. These data have important implications for combining BRAFi/MEKi and checkpoint blockade in the treatment of melanoma.
Collapse
Affiliation(s)
- Zachary A Cooper
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexandre Reuben
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jacob Austin-Breneman
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jennifer A Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
117
|
Queirolo P, Picasso V, Spagnolo F. Combined BRAF and MEK inhibition for the treatment of BRAF-mutated metastatic melanoma. Cancer Treat Rev 2015; 41:519-26. [PMID: 25944484 DOI: 10.1016/j.ctrv.2015.04.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 04/20/2015] [Accepted: 04/22/2015] [Indexed: 02/07/2023]
Abstract
Combined BRAF and MEK inhibition out-performed BRAF inhibitor monotherapy in 3 randomized Phase 3 studies for BRAF-mutated metastatic melanoma patients and the combination of BRAF inhibitor dabrafenib with MEK inhibitor trametinib is now an FDA-approved treatment in this setting. Nevertheless, the majority of patients face progressive disease even when treated with the combination. Mechanisms of resistance to BRAF inhibition have been extensively investigated, whilst less is known about the specific mechanisms of resistance to combined therapy. The aim of this paper is to review the efficacy and safety of the combination of BRAF plus MEK inhibitors compared with BRAF inhibitor monotherapy and immunotherapy, as well as to discuss the existing evidence for the mechanisms of resistance to combined therapy and assess future treatment strategies to improve outcome based on data provided by clinical and translational research studies.
Collapse
Affiliation(s)
- Paola Queirolo
- Department of Medical Oncology, IRCCS San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Virginia Picasso
- Department of Medical Oncology, IRCCS San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Francesco Spagnolo
- Department of Plastic and Reconstructive Surgery, IRCCS San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy.
| |
Collapse
|