Raising the bar: optimizing combinations of targeted therapy and immunotherapy

Similar documents
New Systemic Therapies in Advanced Melanoma

Interleukin-2 Single Agent and Combinations

Disclosure Information. Mary L. Disis

Is There an Optimal Intersection for Targeted and Immunotherapy Treatments for Melanoma?

Releasing the Brakes on Tumor Immunity: Immune Checkpoint Blockade Strategies

Developing Novel Immunotherapeutic Cancer Treatments for Clinical Use

Immunotherapy for the Treatment of Cancer

Outcomes of Patients With Metastatic Melanoma Treated With Immunotherapy Prior to or After BRAF Inhibitors

Priming the Immune System to Kill Cancer and Reverse Tolerance. Dr. Diwakar Davar Assistant Professor, Melanoma and Phase I Therapeutics

Cancer Progress. The State of Play in Immuno-Oncology

Innovations in Immunotherapy - Melanoma. Systemic Therapies October 27, 2018 Charles L. Bane, MD

Immune Checkpoints. PD Dr med. Alessandra Curioni-Fontecedro Department of Hematology and Oncology Cancer Center Zurich University Hospital Zurich

Rationale and results from. BRAFi and immunotherapy

Combination Approaches in Melanoma: A Balancing Act

Immunotherapy in Unresectable or Metastatic Melanoma: Where Do We Stand? Sanjiv S. Agarwala, MD St. Luke s Cancer Center Bethlehem, Pennsylvania

Cancer immunotherapy with oncolytic viruses: more than just lysis

INIBITORE di BRAF nel MELANOMA

BRAF Gene Mutation Testing To Select Melanoma Patients for BRAF Inhibitor Targeted Therapy. Original Policy Date

Corporate Medical Policy

BRAF/MEK inhibitors in the systemic treatment of advanced skin melanoma

DISCLOSURES. Roche/MSD-Merck/Celgene: Research Funding

Determinants of Immunogenicity and Tolerance. Abul K. Abbas, MD Department of Pathology University of California San Francisco

The Immune System. Innate. Adaptive. - skin, mucosal barriers - complement - neutrophils, NK cells, mast cells, basophils, eosinophils

Phase 1 Study Combining Anti-PD-L1 (MEDI4736) With BRAF (Dabrafenib) and/or MEK (Trametinib) Inhibitors in Advanced Melanoma

New paradigms for treating metastatic melanoma

PTAC meeting held on 5 & 6 May (minutes for web publishing)

6/7/16. Melanoma. Updates on immune checkpoint therapies. Molecularly targeted therapies. FDA approval for talimogene laherparepvec (T- VEC)

Personalized medicine - cancer immunotherapy

Immune Checkpoint Inhibitors Drug Combinations: Patients Relevance & Ways Forward

Radiation Therapy as an Immunomodulator

Tumor Immunology: A Primer

Approaches To Treating Advanced Melanoma

Exploring Therapeutic Combinations with anti-ctla-4 Antibody

Novel RCC Targets from Immuno-Oncology and Antibody-Drug Conjugates

2/16/2018. The Immune System and Cancer. Fatal Melanoma Transferred in a Donated Kidney 16 years after Melanoma Surgery

Update on Melanoma Treatment. Tara C Mitchell, MD

Immunotherapy Concept Turned Reality

Best Practices in the Treatment and Management of Metastatic Melanoma. Melanoma

Update on Targeted Therapy in Melanoma

Corporate Medical Policy

Combining targeted therapy and immune checkpoint inhibitors in the treatment of metastatic melanoma

Synergistic combinations of targeted immunotherapy to combat cancer

Immune Checkpoint Inhibitors: The New Breakout Stars in Cancer Treatment

Immunotherapy for the Treatment of Melanoma. Marlana Orloff, MD Thomas Jefferson University Hospital

Immunotherapy versus targeted treatments in metastatic renal cell carcinoma: The return game?

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

Pembrolizumab: First in Class for Treatment of Metastatic Melanoma

Tumor Microenvironment and Immune Suppression

FOCiS. Lecture outline. The immunological equilibrium: balancing lymphocyte activation and control. Immunological tolerance and immune regulation -- 1

Exploring Immunotherapies: Beyond Checkpoint Inhibitors

IMMUNOTHERAPY IN THE TREATMENT OF CERVIX CANCER. Linda Mileshkin, Medical Oncologist Peter MacCallum Cancer Centre, Melbourne Australia

IMMUNOTHERAPY FOR CANCER A NEW HORIZON. Ekaterini Boleti MD, PhD, FRCP Consultant in Medical Oncology Royal Free London NHS Foundation Trust

Lecture outline. Immunological tolerance and immune regulation. Central and peripheral tolerance. Inhibitory receptors of T cells. Regulatory T cells

Is Prostate Cancer Amenable to Immunotherapy Approaches? New Frontiers in Urologic Oncology, September 12, 2015

Molecular mechanisms of the T cellinflamed tumor microenvironment: Implications for cancer immunotherapy

Emerging Targets in Immunotherapy

Normal RAS-RAF (MAPK) pathway signaling

Basic Principles of Tumor Immunotherapy. Ryan J. Sullivan, M.D. Massachusetts General Hospital Cancer Center Boston, MA

National Institute for Health and Care Excellence. Single Technology Appraisal (STA)

Melanoma: Therapeutic Progress and the Improvements Continue

Licia Rivoltini, MD Unit of Immunotherapy of Human Tumors

Terapia Immunomodulante e Target Therapies nel Trattamento del Melanoma Metastatico

Current Trends in Melanoma Theresa Medina, MD UCD Cutaneous Oncology

Review of immunotherapy in melanoma

New therapeutic strategies for BRAF mutant colorectal cancers

Nivolumab in Hodgkin Lymphoma

Combining ADCs with Immuno-Oncology Agents

Tumor Immunity and Immunotherapy. Andrew Lichtman M.D., Ph.D. Brigham and Women s Hospital Harvard Medical School

A Multi-Center Phase II Study on High Dose IL-2 (HD IL-2) Sequenced with Vemurafenib in Patients with BRAF-V600E Mutation Positive Advanced Melanoma

Interaction of Molecular Alterations With Immune Response in Melanoma

Marshall T Bell Research Resident University of Colorado Grand Rounds Nov. 21, 2011

Summary... 2 MELANOMA AND OTHER SKIN TUMOURS... 3

Chapter 7 Conclusions

THE ROLE OF TARGETED THERAPY AND IMMUNOTHERAPY IN THE TREATMENT OF ADVANCED CERVIX CANCER

Is Immune Therapy the Holy Grail in Metastatic Kidney Cancer?

Understanding and overcoming immunoregulatory barriers within

Type 1 Diabetes: Islet expressing GAD65 (green) with DAPI (Blue) Islet expressing Insulin (red) in 3D confocal imaging

Innovation in Prevention, Early Detection & Diagnosis of Colorectal Cancer Heidelberg Workshop Session VI, Oncology Pipeline June 6, 2014

Immunotherapy of patients with metastatic melanoma

Nobel Prize in Physiology or Medicine 2018

Bases for Immunotherapy in Multiple Myeloma

Accepted Manuscript. Next Steps for Immune Checkpoints in Hepatocellular Carcinoma. Patricia M. Santos, Lisa H. Butterfield

Immunotherapie: algemene principes

ONCOS-102 in melanoma Dr. Alexander Shoushtari. 4. ONCOS-102 in mesothelioma 5. Summary & closing

Reviewers' comments: Reviewer #1 (Remarks to the Author):

Cancer immunity and immunotherapy. General principles

A Case Study: Ipilimumab in Pre-treated Metastatic Melanoma

Immunotherapy in non-small cell lung cancer

Immunotherapy, an exciting era!!

Immunotherapy for Malignant Brain Cancer: Overcoming Immune Suppression to Improve the Clinical Experience

Treatment algorithm of metastatic mucosal melanoma

Melanoma: From Chemotherapy to Targeted Therapy and Immunotherapy. What every patient needs to know. James Larkin

T Lymphocyte Activation and Costimulation. FOCiS. Lecture outline

Cover Page. The handle holds various files of this Leiden University dissertation.

Melanoma. Il parere dell esperto. V. Ferraresi. Divisione di Oncologia Medica 1

DEVELOPMENT OF CELLULAR IMMUNOLOGY

Complexity of intra- and inter-pathway loops in colon cancer and melanoma: implications for targeted therapies

Discover the PD-1 pathway and its role in cancer 105/15 -ONCO- 07/15

Darwinian selection and Newtonian physics wrapped up in systems biology

A case report of using nivolumab for a malignant melanoma patient with rheumatoid arthritis

Transcription:

Editorial Page 1 of 5 Raising the bar: optimizing combinations of targeted therapy and immunotherapy Alexandre Reuben 1, Jacob Austin-Breneman 1, Jennifer A. Wargo 1,2 *, Zachary A. Cooper 1,2 * 1 Department of Surgical Oncology; 2 Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA *These authors contributed equally to this work. Correspondence to: Jennifer A. Wargo. Department of Surgical Oncology and Department of Genomic Medicine, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA. Email: jwargo@mdanderson.org. Submitted Oct 15, 2015. Accepted for publication Oct 21, 2015. doi: 10.3978/j.issn.2305-5839.2015.10.28 View this article at: http://dx.doi.org/10.3978/j.issn.2305-5839.2015.10.28 Major breakthroughs have arisen in the treatment of melanoma and other cancers through the use of targeted and immunotherapy. Therapies targeting the BRAF V600E mutation, such as vemurafenib and dabrafenib, were FDAapproved in 2011 and 2013, following demonstration of rapid, marked response in a majority of patients expressing the BRAF V600 mutation and a survival benefit over then standard-of-care therapy with dacarbazine (1,2). However, the vast majority of responding patients eventually relapse, most often within only 6-12 months of treatment initiation (3,4). Another form of immunotherapy, immune checkpoint blockade, exploits a tumor-deployed immune escape mechanism through which tumors impede the immune response by binding checkpoint molecules which serve as brakes, specifically on T lymphocytes. Such therapies involving monoclonal blocking antibodies against cytotoxic T lymphocyte antigen-4 (CTLA-4) and programmed death-1 (PD-1) were approved in 2011 and 2014, respectively. Though these treatments are associated with responses in fewer patients (20-35%) (5,6) than treatment with targeted therapy, responses are often durable (7) with a significant proportion of patients achieving durable disease control. Unfortunately, many patients do not derive benefit from these forms of therapy (1,2,5,6), and more therapeutic options are needed. Another form of therapy that has been studied extensively is adoptive cell therapy (ACT), and involves the isolation and expansion of antigenspecific lymphocytes from tumor (tumor infiltrating lymphocytes-til) (8) or peripheral blood (9) from patients with melanoma (and other cancer types). This form of therapy is associated with responses in approximately 50% of metastatic melanoma patients (10), though its use has been limited by the technical expertise involved in isolation and expansion of these cells, as well as the infrastructure required for this therapeutic approach (11). Given the success and limitations of each of these forms of therapy, there has been great interest in exploring combination strategies incorporating the use of targeted therapy and immunotherapy with several clinical trials incorporating combination approaches currently underway (NCT01940809, NCT01767454, NCT02200562). However complexities exist, as the effect of targeted therapies on host immune cells is not completely understood, and there is evidence that certain targeted agents (e.g., MEK inhibitors) may have deleterious effects on T cells in vitro. Pre-clinical studies deeply examining the mechanism of such approaches are growing in the literature, and will ultimately help inform further combination strategies in patients with melanoma and other cancers. An excellent example of such a paper was recently published in Science Translational Medicine (12). In their paper entitled Improved antitumor activity of immunotherapy with BRAF and MEK inhibitors in BRAF V600E melanoma, Hu-Lieskovan et al. demonstrated superior antitumor effect when dual targeted therapies with BRAF and MEK inhibitors were combined with immunotherapeutic approaches (including ACT and immune checkpoint blockade with anti-pd-1) in a murine model of BRAF V600E mutant metastatic melanoma. Studies such as these are critically important, as

Page 2 of 5 Reuben et al. Improving responses to cancer therapy through combinations targeted therapy agents (such as those targeting BRAF V600E and MEK) succeed in inhibiting crucial signaling and proliferation pathways activated in cancers, but also have unintended effects on the tumor microenvironment, including on immune infiltrates (13). These effects can be advantageous, but at times may also be deleterious. Treatment with BRAF inhibitor therapy is associated with early and significant increases in CD8 + TIL numbers and clonality within tumors of patients with melanoma (14,15), as well as increases in their cytotoxic potential through production of granzyme B and perforin (14). Importantly, these effects may in part be mediated through increases in the expression of human leukocyte antigens (HLAs) (16) and melanoma differentiation antigens (MDAs) gp100, TRP1, TRP2, and MART-1, due to increased transcription of MITF targets as a consequence of MAPK pathway inhibition (14). These favorable changes in the tumor microenvironment are found early during the course of therapy (within the first few weeks) and may be lost later during the course of treatment or at progression on BRAF inhibitors, which has important implications for timing of therapy. Interestingly, BRAF inhibitors manifest paradoxical effects on distinct cell types: they impede proliferation of BRAF V600 cells through MAPK pathway inhibition, while causing MAPK pathway hyperactivation in BRAF WT cells such as T lymphocytes, both in vitro and in vivo (17,18). There was a strong clinical rationale early in testing to empirically combine BRAF-targeted therapy with immunotherapy, in the hopes of achieving high response rates to therapy (such as those observed in targeted therapy) with a longer duration of response (as seen with immunotherapy). Since then, a growing scientific rationale has suggested combining these forms of therapy. A strong example of this is the observation that although BRAF inhibition is associated with a favorable immune microenvironment early in the course of therapy, there is also an increase in the expression of the immunomodulatory ligand for PD-1 (PD-L1) in the tumor microenvironment, suggesting a potential mechanism of adaptive resistance (14,19). Later studies focused on addressing this through combination of BRAF inhibitors and anti-pd-1 therapy in a murine model, and demonstrated improved anti-tumor responses and survival in combination therapy as opposed to monotherapy with either agent, as well as increased TIL number and activation in tumors of mice on combined targeted therapy and PD-1 blockade (15). However, immune effects linked to MEK inhibition have been less well characterized, though there is a growing body of observations suggesting that MEK inhibition of T cells is not substantiated in vivo. This is crucial, as combining MEK inhibition with immunotherapy could prove significant in BRAF WT melanoma as well as in multiple other cancer types, as many of these are RASdriven and could be targeted using MEK inhibitors. This is even relevant in BRAF-mutant melanoma, as the current standard of care treatment for patients with metastatic BRAF-mutant melanoma incorporates combined BRAF and MEK inhibition, based on a survival benefit when compared to BRAF inhibitors alone (20). Multiple in vitro studies have been performed to assess effects of MEK inhibitors (21) and suggest that though there are some beneficial effects, MEK inhibitors may impair T cell function. Beneficial effects include enhanced antigen expression in melanoma, as treatment with MEK inhibitors in BRAF wild-type and BRAF mutant melanoma is associated with increased expression of melanoma antigens (such as MART-1, gp100, TRP1 and TRP2) (22), which could be exploited by administering antigen-specific T lymphocytes via ACT. However, studies have also demonstrated that MEK inhibition may be detrimental to immune cell populations in vitro (22,23). Several studies have shown that treatment with MEK inhibitors leads to impaired T cell proliferation, cytokine secretion, and expansion of antigen-specific T cells (22,23). Importantly, suppressive effects of MEK inhibition were not limited to lymphocytes, as it was demonstrated that MEK inhibition also leads to increased maturation of dendritic cells, resulting in decreased crosspresentation and dampened T cell priming (23,24). MEK inhibition may also affect T cell subsets differentially. In the context of graft versus host disease (GVHD) in the setting of stem cell transplants, it was observed that MEK expression is higher in less differentiated and naive T cells, while decreased in effector memory T cells. As shown by Shindo et al. (21), MEK inhibition preferentially affected cytokine production and reactivity of naive and central memory T cells, while sparing more differentiated effector memory T cells. This work suggests that MEK inhibition has the potential to detrimentally affect the immune system in vitro, but must be further characterized in vivo in order to better understand mechanisms of MEK inhibition and the impact of combination strategies with BRAF inhibition and checkpoint blockade immunotherapy. These issues were elegantly addressed in a manuscript recently published in Science Translational Medicine by Hu-Lieskovan et al. (12). In these studies, the authors

Annals of Translational Medicine, Vol 3, No 18 October 2015 Page 3 of 5 investigated the synergy of BRAF and MEK inhibition with immunotherapeutic regimens such as ACT and checkpoint blockade immunotherapy. The group studied this in the context of melanoma via a murine model, and used CD8 + T cells from transgenic mice that are specific against the melanoma antigen gp100. In these studies, mice were treated with targeted therapy including dabrafenib (BRAF inhibition) and/or trametinib (MEK inhibition), either as targeted therapy alone or in combination with gp100- specific T cells via ACT. It was demonstrated that treatment with dual targeted therapy through combined dabrafenib and trametinib appears to synergize and offer better tumor control than with monotherapy using either agent alone. These studies were extended to combinations with immunotherapy through ACT, and showed total control of tumor burden. Importantly, the efficacy of targeted therapy has been linked to the immune response, which could provide a rationale for the success of combinations with ACT. Therefore, T cells were injected into targeted therapytreated animals and TIL were quantified. Combination of targeted therapy and ACT demonstrated increased homing of T cells to tumors compared to ACT alone within 5 days of treatment, an effect which persisted throughout the course of experiments. Interestingly, the combination of dabrafenib and trametinib presented no improvement in comparison to trametinib alone in enhancing T cell homing to tumors. One of the previous pitfalls linked to using MEK inhibition has been demonstration of decreased proliferation and viability compared to untreated or BRAF inhibitor-treated T cells in vitro. These in vitro results were reproduced by Hu-Lieskovan et al. However, in vivo experiments showed dramatically different findings. Upon injection, it was determined that production of IFN-γ by TIL was unaffected by trametinib treatment. Furthermore, cytotoxicity assays revealed that treatment with dabrafenib and/or trametinib does not impede antigen-specific tumor cytotoxicity, an observation that could be related to differential effects of MEK inhibition on differentiated T cells (12,21). Another immune escape strategy employed by tumors is their induction of immunosuppressive cell populations such as regulatory T cells (Treg). The impact of combination therapies on this population was therefore assessed, and showed that dabrafenib treatment causes an induction of Tregs in tumors within 5 days following treatment initiation, which could likely have inhibitory effects on the anti-tumor response. Importantly, combination of dabrafenib and trametinib reversed these effects, reducing Treg numbers to their control levels and providing further rationale for combining these targeted therapies. In a set of mechanistic studies, transcriptional analysis by microarray was performed and hierarchical clustering demonstrated that dabrafenib and trametinib showed immune signatures associated to chemokine and MHC expression, as well as PD-L1 up-regulation, which could suggest emergence of a resistance mechanism and rationale for combining targeted therapy with PD-1 blockade. Accordingly, Hu-Lieskovan and colleagues also investigated the impact of combining targeted therapy with PD-1 blockade. Previous experiments had suggested that, although ACT and targeted therapy may synergize, this leads to both induction of Tregs within tumors, and expression of PD-L1, potentially as a consequence of increased immune reactivity and IFN-γ production within the tumor microenvironment. In subsequent combination therapies including dual dabrafenib and trametinib targeted therapy and PD-1 checkpoint blockade, this triple combination best controlled tumor growth in vivo, likely through blockade of PD-1/PD-L1 interaction between tumor and T cells. Numerous effective treatment regimens have been developed for patients with metastatic melanoma over the past several years, each with its strengths and weaknesses. However, the vast majority of patients do not achieve durable benefit. These pre-clinical studies suggest that combination strategies may be advantageous and overcome the shortcomings of individual monotherapy approaches. Despite this, several other questions remain. First, can combining targeted and immunotherapies benefit patients in the clinical context, and could this extend to other malignancies? Second, considering the rapid and transient immune response induced by targeted therapies, what is the optimal timing and sequence of combination? Third, can combining targeted and immunotherapy increase (or potentially decrease) toxicity? And lastly, can targeting both essential tumor pathways and immune mechanisms circumvent the established tumor heterogeneity? More work is needed to better understand the mechanisms of synergy between these treatments, as well as how translatable these findings may be to melanoma patients in clinical trials, in the hope of optimizing these therapeutic regimens. Acknowledgements JA Wargo acknowledges the Melanoma Research Alliance

Page 4 of 5 Reuben et al. Improving responses to cancer therapy through combinations Team Science Award, the Kenedy Memorial Foundation grant #0727030 and the generous philanthropic support of several families whose lives have been affected by melanoma. Footnote Provenance: This is a Guest Editorial commissioned by Executive Editor Bing Gu, MD (Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University). Conflicts of Interest: JA Wargo has honoraria from the speakers bureau of Dava Oncology and is an advisory board member for GlaxoSmithKline and Roche/Genentech. References 1. Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med 2011;364:2507-16. 2. Hauschild A, Grob JJ, Demidov LV, et al. Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet 2012;380:358-65. 3. Long GV, Stroyakovskiy D, Gogas H, et al. Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: a multicentre, doubleblind, phase 3 randomised controlled trial. Lancet 2015;386:444-51. 4. McArthur GA, Chapman PB, Robert C, et al. Safety and efficacy of vemurafenib in BRAF(V600E) and BRAF(V600K) mutation-positive melanoma (BRIM-3): extended follow-up of a phase 3, randomised, open-label study. Lancet Oncol 2014;15:323-32. 5. Hodi FS, O'Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010;363:711-23. 6. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-pd-1 antibody in cancer. N Engl J Med 2012;366:2443-54. 7. Sharma P, Allison JP. The future of immune checkpoint therapy. Science 2015;348:56-61. 8. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 2015;348:62-8. 9. Hunder NN, Wallen H, Cao J, et al. Treatment of metastatic melanoma with autologous CD4+ T cells against NY-ESO-1. N Engl J Med 2008;358:2698-703. 10. Rosenberg SA, Yang JC, Sherry RM, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res 2011;17:4550-7. 11. Svane IM, Verdegaal EM. Achievements and challenges of adoptive T cell therapy with tumor-infiltrating or bloodderived lymphocytes for metastatic melanoma: what is needed to achieve standard of care? Cancer Immunol Immunother 2014;63:1081-91. 12. Hu-Lieskovan S, Mok S, Homet Moreno B, et al. Improved antitumor activity of immunotherapy with BRAF and MEK inhibitors in BRAF(V600E) melanoma. Sci Transl Med 2015;7:279ra41. 13. Knight DA, Ngiow SF, Li M, et al. Host immunity contributes to the anti-melanoma activity of BRAF inhibitors. J Clin Invest 2013;123:1371-81. 14. Frederick DT, Piris A, Cogdill AP, et al. BRAF inhibition is associated with enhanced melanoma antigen expression and a more favorable tumor microenvironment in patients with metastatic melanoma. Clin Cancer Res 2013;19:1225-31. 15. Cooper ZA, Juneja VR, Sage PT, et al. Response to BRAF inhibition in melanoma is enhanced when combined with immune checkpoint blockade. Cancer Immunol Res 2014;2:643-54. 16. Bradley SD, Chen Z, Melendez B, et al. BRAFV600E Co-opts a Conserved MHC Class I Internalization Pathway to Diminish Antigen Presentation and CD8+ T-cell Recognition of Melanoma. Cancer Immunol Res 2015;3:602-9. 17. Callahan R, Smith GH. MMTV-induced mammary tumorigenesis: gene discovery, progression to malignancy and cellular pathways. Oncogene 2000;19:992-1001. 18. Koya RC, Mok S, Otte N, et al. BRAF inhibitor vemurafenib improves the antitumor activity of adoptive cell immunotherapy. Cancer Res 2012;72:3928-37. 19. Cooper ZA, Reuben A, Austin-Breneman J, et al. Does It MEK a Difference? Understanding Immune Effects of Targeted Therapy. Clin Cancer Res 2015;21:3102-4. 20. Flaherty KT, Infante JR, Daud A, et al. Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N Engl J Med 2012;367:1694-703. 21. Shindo T, Kim TK, Benjamin CL, et al. MEK inhibitors selectively suppress alloreactivity and graft-versus-host disease in a memory stage-dependent manner. Blood 2013;121:4617-26. 22. Boni A, Cogdill AP, Dang P, et al. Selective BRAFV600E inhibition enhances T-cell recognition of melanoma

Annals of Translational Medicine, Vol 3, No 18 October 2015 Page 5 of 5 without affecting lymphocyte function. Cancer Res 2010;70:5213-9. 23. Vella LJ, Pasam A, Dimopoulos N, et al. MEK inhibition, alone or in combination with BRAF inhibition, affects multiple functions of isolated normal human lymphocytes and dendritic cells. Cancer Immunol Res 2014;2:351-60. 24. Vella LJ, Andrews MC, Pasam A, et al. The kinase inhibitors dabrafenib and trametinib affect isolated immune cell populations. Oncoimmunology 2014;3:e946367. Cite this article as: Reuben A, Austin-Breneman J, Wargo JA, Cooper ZA. Raising the bar: optimizing combinations of targeted therapy and immunotherapy. Ann Transl Med 2015;3(18):272. doi: 10.3978/j.issn.2305-5839.2015.10.28