101
|
Wargo JA, Reuben A, Cooper ZA, Oh KS, Sullivan RJ. Immune Effects of Chemotherapy, Radiation, and Targeted Therapy and Opportunities for Combination With Immunotherapy. Semin Oncol 2015; 42:601-16. [PMID: 26320064 DOI: 10.1053/j.seminoncol.2015.05.007] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
There have been significant advances in cancer treatment over the past several years through the use of chemotherapy, radiation therapy, molecularly targeted therapy, and immunotherapy. Despite these advances, treatments such as monotherapy or monomodality have significant limitations. There is increasing interest in using these strategies in combination; however, it is not completely clear how best to incorporate molecularly targeted and immune-targeted therapies into combination regimens. This is particularly pertinent when considering combinations with immunotherapy, as other types of therapy may have significant impact on host immunity, the tumor microenvironment, or both. Thus, the influence of chemotherapy, radiation therapy, and molecularly targeted therapy on the host anti-tumor immune response and the host anti-host response (ie, autoimmune toxicity) must be taken into consideration when designing immunotherapy-based combination regimens. We present data related to many of these combination approaches in the context of investigations in patients with melanoma and discuss their potential relationship to management of patients with other tumor types. Importantly, we also highlight challenges of these approaches and emphasize the need for continued translational research.
Collapse
Affiliation(s)
- Jennifer A Wargo
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX; Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Alexandre Reuben
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Zachary A Cooper
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX; Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kevin S Oh
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Ryan J Sullivan
- Department of Medical Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA.
| |
Collapse
|
102
|
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: 5.4] [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
|
103
|
Doucette CD, Greenshields AL, Liwski RS, Hoskin DW. Piperine blocks interleukin-2-driven cell cycle progression in CTLL-2 T lymphocytes by inhibiting multiple signal transduction pathways. Toxicol Lett 2015; 234:1-12. [DOI: 10.1016/j.toxlet.2015.01.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 01/26/2015] [Accepted: 01/31/2015] [Indexed: 02/03/2023]
|
104
|
Hu-Lieskovan S, Mok S, Homet Moreno B, Tsoi J, Robert L, Goedert L, Pinheiro EM, Koya RC, Graeber TG, Comin-Anduix B, Ribas A. Improved antitumor activity of immunotherapy with BRAF and MEK inhibitors in BRAF(V600E) melanoma. Sci Transl Med 2015. [PMID: 25787767 DOI: 10.1126/scitranslmed.aaa4691.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Combining immunotherapy and BRAF targeted therapy may result in improved antitumor activity with the high response rates of targeted therapy and the durability of responses with immunotherapy. However, the first clinical trial testing the combination of the BRAF inhibitor vemurafenib and the CTLA4 antibody ipilimumab was terminated early because of substantial liver toxicities. MEK [MAPK (mitogen-activated protein kinase) kinase] inhibitors can potentiate the MAPK inhibition in BRAF mutant cells while potentially alleviating the unwanted paradoxical MAPK activation in BRAF wild-type cells that lead to side effects when using BRAF inhibitors alone. However, there is the concern of MEK inhibitors being detrimental to T cell functionality. Using a mouse model of syngeneic BRAF(V600E)-driven melanoma, SM1, we tested whether addition of the MEK inhibitor trametinib would enhance the antitumor activity of combined immunotherapy with the BRAF inhibitor dabrafenib. Combination of dabrafenib and trametinib with pmel-1 adoptive cell transfer (ACT) showed complete tumor regression, increased T cell infiltration into tumors, and improved in vivo cytotoxicity. Single-agent dabrafenib increased tumor-associated macrophages and T regulatory cells (Tregs) in tumors, which decreased with the addition of trametinib. The triple combination therapy resulted in increased melanosomal antigen and major histocompatibility complex (MHC) expression and global immune-related gene up-regulation. Given the up-regulation of PD-L1 seen with dabrafenib and/or trametinib combined with antigen-specific ACT, we tested the combination of dabrafenib, trametinib, and anti-PD1 therapy in SM1 tumors, and observed superior antitumor effect. Our findings support the testing of triple combination therapy of BRAF and MEK inhibitors with immunotherapy in patients with BRAF(V600E) mutant metastatic melanoma.
Collapse
Affiliation(s)
- Siwen Hu-Lieskovan
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Stephen Mok
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Blanca Homet Moreno
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA. Division of Translational Oncology, Carlos III Health Institute, Madrid 28029, Spain
| | - Jennifer Tsoi
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, CA 90095, USA. Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA 90095, USA
| | - Lidia Robert
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Lucas Goedert
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | | | - Richard C Koya
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Thomas G Graeber
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, CA 90095, USA. Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA 90095, USA. Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095, USA
| | - Begoña Comin-Anduix
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095, USA. Division of Surgical Oncology, Department of Surgery, UCLA, Los Angeles, CA 90095, USA
| | - Antoni Ribas
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA. Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA 90095, USA. Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095, USA. Division of Surgical Oncology, Department of Surgery, UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
105
|
Kim T, Amaria RN, Spencer C, Reuben A, Cooper ZA, Wargo JA. Combining targeted therapy and immune checkpoint inhibitors in the treatment of metastatic melanoma. Cancer Biol Med 2015; 11:237-46. [PMID: 25610709 PMCID: PMC4296084 DOI: 10.7497/j.issn.2095-3941.2014.04.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 10/13/2014] [Indexed: 01/09/2023] Open
Abstract
Melanoma is the deadliest form of skin cancer and has an incidence that is rising faster than any other solid tumor. Metastatic melanoma treatment has considerably progressed in the past five years with the introduction of targeted therapy (BRAF and MEK inhibitors) and immune checkpoint blockade (anti-CTLA4, anti-PD-1, and anti-PD-L1). However, each treatment modality has limitations. Treatment with targeted therapy has been associated with a high response rate, but with short-term responses. Conversely, treatment with immune checkpoint blockade has a lower response rate, but with long-term responses. Targeted therapy affects antitumor immunity, and synergy may exist when targeted therapy is combined with immunotherapy. This article presents a brief review of the rationale and evidence for the potential synergy between targeted therapy and immune checkpoint blockade. Challenges and directions for future studies are also proposed.
Collapse
Affiliation(s)
- Teresa Kim
- 1 Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA ; 2 Harvard Medical School, Boston, MA 02115, USA ; 3 Department of Melanoma Medical Oncology, 4 Department of Genomic Medicine, 5 Department of Surgical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Rodabe N Amaria
- 1 Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA ; 2 Harvard Medical School, Boston, MA 02115, USA ; 3 Department of Melanoma Medical Oncology, 4 Department of Genomic Medicine, 5 Department of Surgical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Christine Spencer
- 1 Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA ; 2 Harvard Medical School, Boston, MA 02115, USA ; 3 Department of Melanoma Medical Oncology, 4 Department of Genomic Medicine, 5 Department of Surgical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Alexandre Reuben
- 1 Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA ; 2 Harvard Medical School, Boston, MA 02115, USA ; 3 Department of Melanoma Medical Oncology, 4 Department of Genomic Medicine, 5 Department of Surgical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Zachary A Cooper
- 1 Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA ; 2 Harvard Medical School, Boston, MA 02115, USA ; 3 Department of Melanoma Medical Oncology, 4 Department of Genomic Medicine, 5 Department of Surgical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer A Wargo
- 1 Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA ; 2 Harvard Medical School, Boston, MA 02115, USA ; 3 Department of Melanoma Medical Oncology, 4 Department of Genomic Medicine, 5 Department of Surgical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
106
|
Liu L, Mayes PA, Eastman S, Shi H, Yadavilli S, Zhang T, Yang J, Seestaller-Wehr L, Zhang SY, Hopson C, Tsvetkov L, Jing J, Zhang S, Smothers J, Hoos A. The BRAF and MEK Inhibitors Dabrafenib and Trametinib: Effects on Immune Function and in Combination with Immunomodulatory Antibodies Targeting PD-1, PD-L1, and CTLA-4. Clin Cancer Res 2015; 21:1639-51. [PMID: 25589619 DOI: 10.1158/1078-0432.ccr-14-2339] [Citation(s) in RCA: 361] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 12/23/2014] [Indexed: 01/12/2023]
Abstract
PURPOSE To assess the immunologic effects of dabrafenib and trametinib in vitro and to test whether trametinib potentiates or antagonizes the activity of immunomodulatory antibodies in vivo. EXPERIMENTAL DESIGN Immune effects of dabrafenib and trametinib were evaluated in human CD4(+) and CD8(+) T cells from healthy volunteers, a panel of human tumor cell lines, and in vivo using a CT26 mouse model. RESULTS Dabrafenib enhanced pERK expression levels and did not suppress human CD4(+) or CD8(+) T-cell function. Trametinib reduced pERK levels, and resulted in partial/transient inhibition of T-cell proliferation/expression of a cytokine and immunomodulatory gene subset, which is context dependent. Trametinib effects were partially offset by adding dabrafenib. Dabrafenib and trametinib in BRAF V600E/K, and trametinib in BRAF wild-type tumor cells induced apoptosis markers, upregulated HLA molecule expression, and downregulated certain immunosuppressive factors such as PD-L1, IL1, IL8, NT5E, and VEGFA. PD-L1 expression in tumor cells was upregulated after acquiring resistance to BRAF inhibition in vitro. Combinations of trametinib with immunomodulators targeting PD-1, PD-L1, or CTLA-4 in a CT26 model were more efficacious than any single agent. The combination of trametinib with anti-PD-1 increased tumor-infiltrating CD8(+) T cells in CT26 tumors. Concurrent or phased sequential treatment, defined as trametinib lead-in followed by trametinib plus anti-PD-1 antibody, demonstrated superior efficacy compared with anti-PD-1 antibody followed by anti-PD-1 plus trametinib. CONCLUSION These findings support the potential for synergy between targeted therapies dabrafenib and trametinib and immunomodulatory antibodies. Clinical exploration of such combination regimens is under way.
Collapse
Affiliation(s)
- Li Liu
- Immuno-Oncology and Combination DPU, GlaxoSmithKline, Collegeville, Pennsylvania
| | - Patrick A Mayes
- Immuno-Oncology and Combination DPU, GlaxoSmithKline, Collegeville, Pennsylvania
| | - Stephen Eastman
- Immuno-Oncology and Combination DPU, GlaxoSmithKline, Collegeville, Pennsylvania
| | - Hong Shi
- Immuno-Oncology and Combination DPU, GlaxoSmithKline, Collegeville, Pennsylvania
| | - Sapna Yadavilli
- Immuno-Oncology and Combination DPU, GlaxoSmithKline, Collegeville, Pennsylvania
| | - Tianqian Zhang
- Immuno-Oncology and Combination DPU, GlaxoSmithKline, Collegeville, Pennsylvania
| | - Jingsong Yang
- Immuno-Oncology and Combination DPU, GlaxoSmithKline, Collegeville, Pennsylvania
| | | | - Shu-Yun Zhang
- Immuno-Oncology and Combination DPU, GlaxoSmithKline, Collegeville, Pennsylvania
| | - Chris Hopson
- Immuno-Oncology and Combination DPU, GlaxoSmithKline, Collegeville, Pennsylvania
| | - Lyuben Tsvetkov
- Immuno-Oncology and Combination DPU, GlaxoSmithKline, Collegeville, Pennsylvania
| | - Junping Jing
- Molecular Medicine Unit, Oncology R&D, GlaxoSmithKline, Collegeville, Pennsylvania
| | - Shu Zhang
- Statistical Science, GlaxoSmithKline, Collegeville, Pennsylvania
| | - James Smothers
- Immuno-Oncology and Combination DPU, GlaxoSmithKline, Collegeville, Pennsylvania
| | - Axel Hoos
- Immuno-Oncology and Combination DPU, GlaxoSmithKline, Collegeville, Pennsylvania.
| |
Collapse
|
107
|
Immune checkpoint modulation: rational design of combination strategies. Pharmacol Ther 2015; 150:23-32. [PMID: 25583297 DOI: 10.1016/j.pharmthera.2015.01.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 01/05/2015] [Indexed: 12/11/2022]
Abstract
Immune recognition and elimination of malignant cells require a series of steps orchestrated by the innate and the adaptive arms of the immune system. The majority of tumors have evolved mechanisms that allow for successful evasion of these immune responses. Recognition of these evasive processes led to the development of immunotherapeutic antibodies targeting the co-stimulatory and co-inhibitory receptors on T cells, with the goal of enhancement of T cell activation or reversal of tumor-induced T cell inhibition. Several of these agents, such as antibodies targeting cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed death receptor 1 (PD-1) have already demonstrated significant promise in clinical trials. Clinical benefit of these antibodies as single agents, however, has been limited to a subset of patients and has not been observed in all tumor types. These limitations call for the development of rational combination strategies aiming to extend therapeutic benefit to a broader range of patients. These include: 1) modalities that enhance antigen presentation, such as radiation, cryotherapy, chemotherapy, targeted agents, vaccines, toll-like receptor (TLR) agonists, type I interferon, and oncolytic viruses; 2) additional agents aiming to reverse T cell dysfunction, such as other immune checkpoint inhibitors; and 3) agents targeting other immune inhibitory mechanisms, such as inhibitors of indoleamine dioxygenase (IDO), regulatory T cells, and myeloid-derived suppressor cells (MDSCs). It is becoming increasingly evident that the efficacy of specific combinations will likely not be universal and that the choice of a treatment modality may need to be tailored to fit the needs of each individual patient.
Collapse
|
108
|
Reuben A, Amaria RN, Cooper ZA, Wargo JA. RAF Inhibitor Therapy Promotes Melanocytic Antigen Expression and Enhanced Anti-Tumor Immunity in Melanoma. JOURNAL OF PIGMENTARY DISORDERS 2014; 1. [PMID: 29250602 PMCID: PMC5731654 DOI: 10.4172/2376-0427.1000139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Melanoma remains a major cause of morbidity and mortality worldwide, however tremendous advances have been made in its treatment over the past several years. The discovery of genomic alterations that contribute to oncogenicity has ushered in a new era of molecularly-targeted therapy. Importantly, over half of melanomas harbor a mutation in the BRAF gene that leads to constitutive signaling down the MAPK pathway and multiple subsequent deleterious effects. Pharmacologic agents targeting this mutation have been developed and several are now FDA-approved, having yielded high response rates to therapy although these are tempered by a short duration of response. Multiple molecular mechanisms of resistance have been identified; however until recently few studies had delved into the immune effects of BRAF inhibitors. The effect of BRAF inhibition on anti-tumor immunity will be discussed herein, as will potential implications of these findings in the treatment of melanoma.
Collapse
Affiliation(s)
- Alexandre Reuben
- Division of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Rodabe N Amaria
- Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Zachary A Cooper
- Division of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Jennifer A Wargo
- Division of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
109
|
Vella LJ, Andrews MC, Pasam A, Woods K, Behren A, Cebon JS. The kinase inhibitors dabrafenib and trametinib affect isolated immune cell populations. Oncoimmunology 2014; 3:e946367. [PMID: 25610732 DOI: 10.4161/21624011.2014.946367] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 06/13/2014] [Indexed: 11/19/2022] Open
Abstract
Metastatic melanoma is frequently fatal. Optimal treatment regimens require both rapid and durable disease control, likely best achieved by combining targeted agents with immunotherapeutics. In order to accomplish this, a detailed understanding of the immune consequences of the kinase inhibitors used to treat melanoma is required.
Collapse
Affiliation(s)
- Laura J Vella
- Ludwig Institute for Cancer Research; Melbourne-Austin Branch; Cancer Immuno-biology Laboratory ; Heidelberg, Australia
| | - Miles C Andrews
- Ludwig Institute for Cancer Research; Melbourne-Austin Branch; Cancer Immuno-biology Laboratory ; Heidelberg, Australia
| | - Anupama Pasam
- Ludwig Institute for Cancer Research; Melbourne-Austin Branch; Cancer Immuno-biology Laboratory ; Heidelberg, Australia
| | - Katherine Woods
- Ludwig Institute for Cancer Research; Melbourne-Austin Branch; Cancer Immuno-biology Laboratory ; Heidelberg, Australia
| | - Andreas Behren
- Ludwig Institute for Cancer Research; Melbourne-Austin Branch; Cancer Immuno-biology Laboratory ; Heidelberg, Australia
| | - Jonathan S Cebon
- Ludwig Institute for Cancer Research; Melbourne-Austin Branch; Cancer Immuno-biology Laboratory ; Heidelberg, Australia
| |
Collapse
|
110
|
Vella LJ, Andrews MC, Behren A, Cebon J, Woods K. Immune consequences of kinase inhibitors in development, undergoing clinical trials and in current use in melanoma treatment. Expert Rev Clin Immunol 2014; 10:1107-23. [PMID: 24939732 DOI: 10.1586/1744666x.2014.929943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Metastatic malignant melanoma is a frequently fatal cancer. In recent years substantial therapeutic progress has occurred with the development of targeted kinase inhibitors and immunotherapeutics. Targeted therapies often result in rapid clinical benefit however responses are seldom durable. Immune therapies can result in durable disease control but responses may not be immediate. Optimal cancer therapy requires both rapid and durable cancer control and this can likely best be achieved by combining targeted therapies with immunotherapeutics. To achieve this, a detailed understanding of the immune consequences of the various kinase inhibitors, in development, clinical trial and currently used to treat melanoma is required.
Collapse
Affiliation(s)
- Laura J Vella
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Cancer Immuno-biology Laboratory, Heidelberg, VIC 3084, Australia
| | | | | | | | | |
Collapse
|
111
|
Diwakar D, Choi T, Tawbi TH, Kirkwood JM. Re: Differential influence of vemurafenib and dabrafenib on patients' lymphocytes despite similar clinical efficacy in melanoma. Ann Oncol 2014; 25:1670-1. [PMID: 24890848 DOI: 10.1093/annonc/mdu196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- D Diwakar
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, USA
| | - T Choi
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, USA
| | - T H Tawbi
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, USA
| | - J M Kirkwood
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, USA
| |
Collapse
|