1901
|
Kranz LM, Birtel M, Krienke C, Grunwitz C, Petschenka J, Reuter KC, van de Roemer N, Vascotto F, Vormehr M, Kreiter S, Diken M. CIMT 2015: The right patient for the right therapy - Report on the 13th annual meeting of the Association for Cancer Immunotherapy. Hum Vaccin Immunother 2015; 12:213-21. [PMID: 26186022 PMCID: PMC4962731 DOI: 10.1080/21645515.2015.1068485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 06/29/2015] [Indexed: 12/22/2022] Open
Affiliation(s)
- Lena M Kranz
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH; Mainz, Germany
- Research Center for Immunotherapy (FZI); University Medical Center; Johannes Gutenberg University; Mainz, Germany
| | - Matthias Birtel
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH; Mainz, Germany
| | - Christina Krienke
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH; Mainz, Germany
- Research Center for Immunotherapy (FZI); University Medical Center; Johannes Gutenberg University; Mainz, Germany
| | - Christian Grunwitz
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH; Mainz, Germany
- BioNTech RNA Pharmaceuticals GmbH; Mainz, Germany
| | - Jutta Petschenka
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH; Mainz, Germany
| | | | - Niels van de Roemer
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH; Mainz, Germany
- Research Center for Immunotherapy (FZI); University Medical Center; Johannes Gutenberg University; Mainz, Germany
| | - Fulvia Vascotto
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH; Mainz, Germany
| | - Mathias Vormehr
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH; Mainz, Germany
- BioNTech RNA Pharmaceuticals GmbH; Mainz, Germany
| | - Sebastian Kreiter
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH; Mainz, Germany
| | - Mustafa Diken
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH; Mainz, Germany
| |
Collapse
|
1902
|
McArthur GA. Combination Therapies to Inhibit the RAF/MEK/ERK Pathway in Melanoma: We are not Done Yet. Front Oncol 2015; 5:161. [PMID: 26236691 PMCID: PMC4505146 DOI: 10.3389/fonc.2015.00161] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Accepted: 07/03/2015] [Indexed: 12/19/2022] Open
Affiliation(s)
- Grant A McArthur
- Department of Cancer Medicine, Peter MacCallum Cancer Centre , East Melbourne, VIC , Australia ; Department of Pathology, University of Melbourne , Parkville, VIC , Australia ; Department of Medicine, St Vincent's Hospital, University of Melbourne , Fitzroy, VIC , Australia ; Sir Peter MacCallum Department of Oncology, University of Melbourne , East Melbourne, VIC , Australia ; Molecular Oncology Laboratory, Oncogenic Signaling and Growth Control Program, Peter MacCallum Cancer Centre , East Melbourne, VIC , Australia ; Translational Research Laboratory, Cancer Therapeutics Program, Peter MacCallum Cancer Centre , East Melbourne, VIC , Australia
| |
Collapse
|
1903
|
Abstract
Anaplastic thyroid carcinoma (ATC) is a rare, poorly differentiated type of thyroid cancer occurring in less than 5% of all thyroid cancers. Patients typically have a poor prognosis with very few options for treatment. (2) With current therapy of surgery, chemotherapy, and radiation, median survival is only 6 months from the time of diagnosis. Several mutations in cell cycle regulation have been discovered in ATC that contribute to its undifferentiated state, one of which is the BRAF kinase mutation. This mutation results in activation of the MAPK pathway and uncontrolled cell proliferation. In this case report, a 51 y old male presented with a 2-week history of hoarseness and was diagnosed with ATC. Genetic analysis revealed a mutation in BRAF kinase; the patient subsequently began therapy with vemurafenib, a BRAF kinase inhibitor indicated for melanoma. After an initial response, the patient quickly declined and consequently died from his disease. Anaplastic thyroid carcinoma is a deadly cancer without an effective treatment. Inhibiting mutated enzymes that drive the development of this cancer is a potential drug target that may improve outcomes in patients with ATC.
Collapse
Affiliation(s)
| | - Vinay K Gudena
- b University Hospitals Seidman Cancer Center ; Cleveland , OH USA
| |
Collapse
|
1904
|
Verduzco D, Flaherty KT, Smalley KSM. Feeling energetic? New strategies to prevent metabolic reprogramming in melanoma. Exp Dermatol 2015; 24:657-8. [PMID: 26010519 DOI: 10.1111/exd.12763] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2015] [Indexed: 01/09/2023]
Affiliation(s)
- Daniel Verduzco
- The Department of Tumor Biology, The Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Keith T Flaherty
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Keiran S M Smalley
- The Department of Tumor Biology, The Moffitt Cancer Center & Research Institute, Tampa, FL, USA.,The Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| |
Collapse
|
1905
|
Hernandez-Davies JE, Tran TQ, Reid MA, Rosales KR, Lowman XH, Pan M, Moriceau G, Yang Y, Wu J, Lo RS, Kong M. Vemurafenib resistance reprograms melanoma cells towards glutamine dependence. J Transl Med 2015; 13:210. [PMID: 26139106 PMCID: PMC4490757 DOI: 10.1186/s12967-015-0581-2] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 06/24/2015] [Indexed: 12/13/2022] Open
Abstract
Background V600BRAF mutations drive approximately 50% of metastatic melanoma which can be therapeutically targeted by BRAF inhibitors (BRAFi) and, based on resistance mechanisms, the combination of BRAF and MEK inhibitors (BRAFi + MEKi). Although the combination therapy has been shown to provide superior clinical benefits, acquired resistance is still prevalent and limits the overall survival benefits. Recent work has shown that oncogenic changes can lead to alterations in tumor cell metabolism rendering cells addicted to nutrients, such as the amino acid glutamine. Here, we evaluated whether melanoma cells with acquired resistance display glutamine dependence and whether glutamine metabolism can be a potential molecular target to treat resistant cells. Methods Isogenic BRAFi sensitive parental V600BRAF mutant melanoma cell lines and resistant (derived by chronic treatment with vemurafenib) sub-lines were used to assess differences in the glutamine uptake and sensitivity to glutamine deprivation. To evaluate a broader range of resistance mechanisms, isogenic pairs where the sub-lines were resistant to BRAFi + MEKi were also studied. Since resistant cells demonstrated increased sensitivity to glutamine deficiency, we used glutaminase inhibitors BPTES [bis-2-(5 phenylacetamido-1, 2, 4-thiadiazol-2-yl) ethyl sulfide] and L–L-DON (6-Diazo-5-oxo-l-norleucine) to treat MAPK pathway inhibitor (MAPKi) resistant cell populations both in vitro and in vivo. Results We demonstrated that MAPKi-acquired resistant cells uptook greater amounts of glutamine and have increased sensitivity to glutamine deprivation than their MAPKi-sensitive counterparts. In addition, it was found that both BPTES and L-DON were more effective at decreasing cell survival of MAPKi-resistant sub-lines than parental cell populations in vitro. We also showed that mutant NRAS was critical for glutamine addiction in mutant NRAS driven resistance. When tested in vivo, we found that xenografts derived from resistant cells were more sensitive to BPTES or L-DON treatment than those derived from parental cells. Conclusion Our study is a proof-of-concept for the potential of targeting glutamine metabolism as an alternative strategy to suppress acquired MAPKi-resistance in melanoma.
Collapse
Affiliation(s)
- Jenny E Hernandez-Davies
- Department of Cancer Biology, Beckman Research Institute of City of Hope Cancer Center, Duarte, CA, 91010, USA.
| | - Thai Q Tran
- Department of Cancer Biology, Beckman Research Institute of City of Hope Cancer Center, Duarte, CA, 91010, USA.
| | - Michael A Reid
- Department of Cancer Biology, Beckman Research Institute of City of Hope Cancer Center, Duarte, CA, 91010, USA.
| | - Kimberly R Rosales
- Department of Cancer Biology, Beckman Research Institute of City of Hope Cancer Center, Duarte, CA, 91010, USA.
| | - Xazmin H Lowman
- Department of Cancer Biology, Beckman Research Institute of City of Hope Cancer Center, Duarte, CA, 91010, USA.
| | - Min Pan
- Department of Cancer Biology, Beckman Research Institute of City of Hope Cancer Center, Duarte, CA, 91010, USA.
| | - Gatien Moriceau
- Division of Dermatology/Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1662, USA. .,Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1662, USA.
| | - Ying Yang
- Department of Cancer Biology, Beckman Research Institute of City of Hope Cancer Center, Duarte, CA, 91010, USA.
| | - Jun Wu
- Animal Tumor Model Program, Division of Comparative Medicine, Beckman Research Institute of City of Hope Cancer Center, Duarte, CA, 91010, USA.
| | - Roger S Lo
- Division of Dermatology/Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1662, USA. .,Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-1662, USA.
| | - Mei Kong
- Department of Cancer Biology, Beckman Research Institute of City of Hope Cancer Center, Duarte, CA, 91010, USA.
| |
Collapse
|
1906
|
Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, Schadendorf D, Dummer R, Smylie M, Rutkowski P, Ferrucci PF, Hill A, Wagstaff J, Carlino MS, Haanen JB, Maio M, Marquez-Rodas I, McArthur GA, Ascierto PA, Long GV, Callahan MK, Postow MA, Grossmann K, Sznol M, Dreno B, Bastholt L, Yang A, Rollin LM, Horak C, Hodi FS, Wolchok JD. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N Engl J Med 2015; 373:23-34. [PMID: 26027431 PMCID: PMC5698905 DOI: 10.1056/nejmoa1504030] [Citation(s) in RCA: 6149] [Impact Index Per Article: 614.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Nivolumab (a programmed death 1 [PD-1] checkpoint inhibitor) and ipilimumab (a cytotoxic T-lymphocyte-associated antigen 4 [CTLA-4] checkpoint inhibitor) have been shown to have complementary activity in metastatic melanoma. In this randomized, double-blind, phase 3 study, nivolumab alone or nivolumab plus ipilimumab was compared with ipilimumab alone in patients with metastatic melanoma. METHODS We assigned, in a 1:1:1 ratio, 945 previously untreated patients with unresectable stage III or IV melanoma to nivolumab alone, nivolumab plus ipilimumab, or ipilimumab alone. Progression-free survival and overall survival were coprimary end points. Results regarding progression-free survival are presented here. RESULTS The median progression-free survival was 11.5 months (95% confidence interval [CI], 8.9 to 16.7) with nivolumab plus ipilimumab, as compared with 2.9 months (95% CI, 2.8 to 3.4) with ipilimumab (hazard ratio for death or disease progression, 0.42; 99.5% CI, 0.31 to 0.57; P<0.001), and 6.9 months (95% CI, 4.3 to 9.5) with nivolumab (hazard ratio for the comparison with ipilimumab, 0.57; 99.5% CI, 0.43 to 0.76; P<0.001). In patients with tumors positive for the PD-1 ligand (PD-L1), the median progression-free survival was 14.0 months in the nivolumab-plus-ipilimumab group and in the nivolumab group, but in patients with PD-L1-negative tumors, progression-free survival was longer with the combination therapy than with nivolumab alone (11.2 months [95% CI, 8.0 to not reached] vs. 5.3 months [95% CI, 2.8 to 7.1]). Treatment-related adverse events of grade 3 or 4 occurred in 16.3% of the patients in the nivolumab group, 55.0% of those in the nivolumab-plus-ipilimumab group, and 27.3% of those in the ipilimumab group. CONCLUSIONS Among previously untreated patients with metastatic melanoma, nivolumab alone or combined with ipilimumab resulted in significantly longer progression-free survival than ipilimumab alone. In patients with PD-L1-negative tumors, the combination of PD-1 and CTLA-4 blockade was more effective than either agent alone. (Funded by Bristol-Myers Squibb; CheckMate 067 ClinicalTrials.gov number, NCT01844505.).
Collapse
Affiliation(s)
- James Larkin
- Department of Medical Oncology, Royal Marsden Hospital, London, United Kingdom
| | | | - Rene Gonzalez
- Division of Medical Oncology, University of Colorado Denver, Denver, CO
| | - Jean Jacques Grob
- Aix-Marseille University, Hôpital de La Timone APHM, Marseille, France
| | | | - Christopher D. Lao
- Departments of Internal Medicine and Dermatology, University of Michigan, Ann Arbor, MI
| | | | | | | | - Piotr Rutkowski
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | | | - Andrew Hill
- Tasman Oncology Research, Southport Gold Coast, Queensland, Australia
| | - John Wagstaff
- South West Wales Cancer Institute, Singleton Hospital, Swansea, United Kingdom
| | - Matteo S. Carlino
- Westmead and Blacktown Hospitals, Melanoma Institute Australia and the University of Sydney, Sydney, Australia
| | - John B. Haanen
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Ivan Marquez-Rodas
- Servicio de Oncología Médica, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Grant A. McArthur
- Peter MacCallum Cancer Centre and University of Melbourne, Victoria, Australia
| | | | - Georgina V. Long
- Melanoma Institute Australia, the University of Sydney, and the Mater Hospital, Sydney, Australia
| | - Margaret K. Callahan
- Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - Michael A. Postow
- Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | | | - Mario Sznol
- Yale Cancer Center, Smilow Cancer Hospital of the Yale-New Haven Hospital, Yale University School of Medicine, New Haven, CT
| | | | - Lars Bastholt
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | | | | | | | | | - Jedd D. Wolchok
- Ludwig Center at Memorial Sloan Kettering Cancer Center and Weill-Cornell Medical College, New York, NY
| |
Collapse
|
1907
|
The Role of Regional Therapies for in-Transit Melanoma in the Era of Improved Systemic Options. Cancers (Basel) 2015; 7:1154-77. [PMID: 26140669 PMCID: PMC4586763 DOI: 10.3390/cancers7030830] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Revised: 06/17/2015] [Accepted: 06/24/2015] [Indexed: 12/21/2022] Open
Abstract
The incidence of melanoma has been increasing at a rapid rate, with 4%–11% of all melanoma recurrences presenting as in-transit disease. Treatments for in-transit melanoma of the extremity are varied and include surgical excision, lesional injection, regional techniques and systemic therapies. Excision to clear margins is preferred; however, in cases of widespread disease, this may not be practical. Historically, intralesional therapies were generally not curative and were often used for palliation or as adjuncts to other therapies, but recent advances in oncolytic viruses may change this paradigm. Radiation as a regional therapy can be quite locally toxic and is typically relegated to disease control and symptom relief in patients with limited treatment options. Regional therapies such as isolated limb perfusion and isolated limb infusion are older therapies, but offer the ability to treat bulky disease for curative intent with a high response rate. These techniques have their associated toxicities and can be technically challenging. Historically, systemic therapy with chemotherapies and biochemotherapies were relatively ineffective and highly toxic. With the advent of novel immunotherapeutic and targeted small molecule agents for the treatment of metastatic melanoma, the armamentarium against in-transit disease has expanded. Given the multitude of options, many different combinations and sequences of therapies can be offered to patients with in-transit extremity melanoma in the contemporary era. Reported response and survival rates of the varied treatments may offer valuable information regarding treatment decisions for patients with in-transit melanoma and provide rationale for these decisions.
Collapse
|
1908
|
Shelledy L, Roman D. Vemurafenib: First-in-Class BRAF-Mutated Inhibitor for the Treatment of Unresectable or Metastatic Melanoma. J Adv Pract Oncol 2015; 6:361-5. [PMID: 26705496 PMCID: PMC4677808 DOI: 10.6004/jadpro.2015.6.4.6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
| | - Danielle Roman
- West Penn Allegheny Oncology Network, Pittsburgh, Pennsylvania
| |
Collapse
|
1909
|
Zhao G, Li Q, Wang A, Jiao J. YY1 regulates melanoma tumorigenesis through a miR-9 ~ RYBP axis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:66. [PMID: 26104682 PMCID: PMC4511530 DOI: 10.1186/s13046-015-0177-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 05/27/2015] [Indexed: 12/22/2022]
Abstract
Background The Yin Yang 1 (YY1) transcription factor has been identified to target a plethora of potential target genes, which are important for cell proliferation and differentiation. Although the role that YY1 plays in different human types of cancer has been reported, its biological and mechanistic significance in melanoma has not been well defined. Methods Quantitative RT-PCR analysis was used to determine whether aberrant YY1 and miR-9 expression occurred in melanoma, compared with benign nevi and normal tissue controls. Furthermore, the transcriptional regulation of YY1 on miR-9 expression was assessed by using quantitative ChIP-PCR assay. Subsequently, the effects of YY1 and miR-9 on proliferation, cell cycle, migration and invasion of melanoma cells were detected using CCK-8, flow cytometric analysis, wound healing and transwell invasion assays, respectively. Finally, the post-transcriptional regulation of miR-9 on RYBP was analyzed using luciferase reporter and immunoblot analysis. Results Elevated YY1 levels were observed in patients with melanoma, compared with benign nevi and normal tissue controls, and the increased YY1 was associated with melanoma metastasis state and tumor stage. Furthermore, YY1 negatively regulated miR-9 transcription. Silencing of YY1 inhibited proliferation, cell cycle progression, migration and invasion in melanoma cells, while ectopic of miR-9 did the same. Additionally, RYBP was shown to be a direct target of miR-9 through binding to its 3′ UTR, thus forming a YY1 ~ miR-9 ~ RYBP axis. Conclusions These results identify a novel YY1 ~ miR-9 ~ RYBP axis involved in melanoma tumorigenesis and reinforce the idea that regulatory circuitries involving miRNAs and TFs are prevalent mechanisms.
Collapse
Affiliation(s)
- Guowei Zhao
- Department of Dermatology, The Central Hospital of Zibo City, Zibo, 255036, Shandong Province, China.
| | - Qiang Li
- Oncology Department, The Foruth People's Hospital of Zibo City, Zibo, 255067, Shandong Province, China.
| | - Aiqin Wang
- Department of Dermatology, The Central Hospital of Zibo City, Zibo, 255036, Shandong Province, China.
| | - Jian Jiao
- Department of Dermatology, Qilu Hospital Shandong University, 107 Wenhuaxi Road, Jinan, 250012, Shandong Province, China.
| |
Collapse
|
1910
|
Minor DR, Puzanov I, Callahan MK, Hug BA, Hoos A. Severe gastrointestinal toxicity with administration of trametinib in combination with dabrafenib and ipilimumab. Pigment Cell Melanoma Res 2015; 28:611-2. [PMID: 25996827 PMCID: PMC4744965 DOI: 10.1111/pcmr.12383] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Affiliation(s)
- David R Minor
- California Pacific Center for Melanoma Research and Treatment, San Francisco, CA, USA
| | - Igor Puzanov
- Division of Hematology-Oncology, Vanderbilt University, Nashville, TN, USA
| | | | - Bruce A Hug
- Oncology Research and Development, Glaxo Smith Kline, Collegeville, PA, USA
| | - Axel Hoos
- Oncology Research and Development, Glaxo Smith Kline, Collegeville, PA, USA
| |
Collapse
|
1911
|
Johnson DB, Puzanov I, Kelley MC. Talimogene laherparepvec (T-VEC) for the treatment of advanced melanoma. Immunotherapy 2015; 7:611-9. [PMID: 26098919 DOI: 10.2217/imt.15.35] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Melanoma often spreads to cutaneous or subcutaneous sites that are amenable to direct, intralesional injection. As such, developing effective injectable agents has been of considerable interest. Talimogene laherperepvec (T-VEC) is an injectable modified oncolytic herpes virus being developed for the treatment of advanced melanoma. Pre-clinical studies have shown that T-VEC preferentially infects melanoma cells and exerts antitumor activity through directly mediating cell death and by augmenting local and even distant immune responses. T-VEC has now been assessed in Phase II and III clinical trials and has demonstrated a tolerable side-effect profile and promising efficacy, showing an improved durable response rate and a trend toward superior overall survival compared to granulocyte-macrophage colony-stimulating factor. Despite these promising results, responses have been uncommon in patients with visceral metastases. T-VEC is currently being evaluated in combination with other immune therapies (ipilimumab and pembrolizumab) with early signs of activity. In this review, we discuss the preclinical rationale, the clinical experience, and future directions for T-VEC in advanced melanoma.
Collapse
Affiliation(s)
- Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, 777 PRB, 2220 Pierce Ave, Nashville, TN 37232, USA
| | - Igor Puzanov
- Department of Medicine, Vanderbilt University Medical Center, 777 PRB, 2220 Pierce Ave, Nashville, TN 37232, USA
| | - Mark C Kelley
- Department of Surgery, Vanderbilt University Medical Center, TN, USA
| |
Collapse
|
1912
|
He M, Lovell J, Ng BL, Spillane J, Speakman D, Henderson MA, Shackleton M, Gyorki DE. Post-operative survival following metastasectomy for patients receiving BRAF inhibitor therapy is associated with duration of pre-operative treatment and elective indication. J Surg Oncol 2015; 111:980-4. [PMID: 26080731 DOI: 10.1002/jso.23938] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 04/30/2015] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Metastasectomy can provide durable disease control for selected patients with metastatic melanoma. Vemurafenib is a BRAF kinase inhibitor which has demonstrated significant improvement in disease-specific survival in patients with metastatic melanoma with a BRAF gene mutation. This study examined the efficacy and safety of metastasectomy during treatment with vemurafenib. METHODS A retrospective review was performed of all patients receiving vemurafenib at Peter MacCallum Cancer Centre. Patient records were reviewed to identify patients undergoing surgery within 30 days of vemurafenib therapy. Descriptive statistics and survival analysis were performed. RESULTS Nineteen patients underwent 21 metastasectomies including craniotomy (57%), spinal decompression (14%), small bowel resection (14%), lung resection (9.5%) and neck dissection (4.5%). Indications for surgery were: an isolated residual focus of disease (n = 2); isolated progressive disease in the setting of stability elsewhere (n = 9); and symptomatic disease (n = 8). Grade 2 or higher surgical complications occurred in 19% of cases and there was one peri-operative death. Median post-operative survival was seven months. There was a trend toward improved post-operative survival for patients with longer duration of vemurafenib therapy (P = 0.04) and for those undergoing elective surgery (P = 0.07). CONCLUSION Resection of oligometastatic disease during BRAF-targeted therapy is safe. Selected patients have durable post-operative disease control.
Collapse
Affiliation(s)
- Mike He
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Jane Lovell
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Bee Ling Ng
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - John Spillane
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - David Speakman
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Michael A Henderson
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, East Melbourne, Australia.,Department of Surgery, University of Melbourne, Parkville, Australia
| | - Mark Shackleton
- Department of Cancer Medicine, Peter MacCallum Cancer Centre, East Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - David E Gyorki
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, East Melbourne, Australia.,Department of Surgery, University of Melbourne, Parkville, Australia
| |
Collapse
|
1913
|
Sweetlove M, Wrightson E, Kolekar S, Rewcastle GW, Baguley BC, Shepherd PR, Jamieson SMF. Inhibitors of pan-PI3K Signaling Synergize with BRAF or MEK Inhibitors to Prevent BRAF-Mutant Melanoma Cell Growth. Front Oncol 2015; 5:135. [PMID: 26137449 PMCID: PMC4468830 DOI: 10.3389/fonc.2015.00135] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/01/2015] [Indexed: 11/13/2022] Open
Abstract
BRAF and MEK inhibitors have improved outcomes for patients with BRAF-mutant melanoma, but their efficacy is limited by both intrinsic and acquired resistances. Activation of the PI3K pathway can mediate resistance to these agents, providing a strong rationale for combination therapy in melanoma. Here, a panel of nine low-passage human metastatic melanoma cell lines with BRAF mutations was tested in cell proliferation and protein expression assays for sensitivity to inhibitors of MEK (selumetinib) and BRAF (vemurafenib) as single agents and in combination with inhibitors of pan-PI3K (ZSTK474), pan-PI3K/mTOR (BEZ235), individual PI3K isoforms (p110α, A66; p110β, TGX-221; p110γ, AS-252424; p110δ, idelalisib), or mTORC1/2 (KU-0063794). Selumetinib and vemurafenib potently inhibited cell proliferation in all cell lines, especially in those that expressed low levels of phosphorylated AKT (pAKT). ZSTK474 and BEZ235 also inhibited cell proliferation in all cell lines and enhanced the antitumor activity of selumetinib and vemurafenib in the majority of lines by either interacting synergistically or additively to increase potency or by inducing cytotoxicity by significantly increasing the magnitude of cell growth inhibition. Furthermore, ZSTK474 or BEZ235 combined with selumetinib to produce robust inhibition of pERK, pAKT, and pS6 expression and synergistic inhibition of NZM20 tumor growth. The inhibitors of individual PI3K isoforms or mTORC1/2 were less effective at inhibiting cell proliferation either as single agents or in combination with selumetinib or vemurafenib, although KU-0063794 synergistically interacted with vemurafenib and increased the magnitude of cell growth inhibition with selumetinib or vemurafenib in certain cell lines. Overall, these results suggest that the sensitivity of BRAF-mutant melanoma cells to BRAF or MEK inhibitors is at least partly mediated by activation of the PI3K pathway and can be enhanced by combined inhibition of the BRAF/MEK and PI3K/mTOR signaling pathways.
Collapse
Affiliation(s)
- Melanie Sweetlove
- Auckland Cancer Society Research Centre, The University of Auckland , Auckland , New Zealand
| | - Emma Wrightson
- Auckland Cancer Society Research Centre, The University of Auckland , Auckland , New Zealand
| | - Sharada Kolekar
- Department of Molecular Medicine and Pathology, The University of Auckland , Auckland , New Zealand
| | - Gordon W Rewcastle
- Auckland Cancer Society Research Centre, The University of Auckland , Auckland , New Zealand ; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland , Auckland , New Zealand
| | - Bruce C Baguley
- Auckland Cancer Society Research Centre, The University of Auckland , Auckland , New Zealand ; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland , Auckland , New Zealand
| | - Peter R Shepherd
- Department of Molecular Medicine and Pathology, The University of Auckland , Auckland , New Zealand ; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland , Auckland , New Zealand
| | - Stephen M F Jamieson
- Auckland Cancer Society Research Centre, The University of Auckland , Auckland , New Zealand ; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland , Auckland , New Zealand
| |
Collapse
|
1914
|
Bertotti A, Sassi F. Molecular Pathways: Sensitivity and Resistance to Anti-EGFR Antibodies. Clin Cancer Res 2015; 21:3377-83. [DOI: 10.1158/1078-0432.ccr-14-0848] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 04/28/2015] [Indexed: 11/16/2022]
|
1915
|
Orloff M, Valsecchi ME, Sato T. Successes and setbacks of early investigational drugs for melanoma. Expert Opin Investig Drugs 2015; 24:993-7. [PMID: 26068553 DOI: 10.1517/13543784.2015.1051618] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Treatment of advanced and metastatic melanoma is a rapidly changing field. Over the past 10 years, there have been six new drugs approved by the FDA for the treatment of metastatic melanoma. These approved drugs include a number of immune checkpoint inhibitors and MAPK-pathway-targeted therapies. The discovery of such agents as ipilimumab, pembrolizumab, nivolumab, vemurafenib, trametininb and dabrafenib have revolutionized the way in which melanoma in managed. While these agents have succeeded in both early and later phase clinical trials, a large number of investigational therapies have not yet been developed or researched past Phase I clinical studies. Furthermore, there are dozens of potential agents in Phase I and Phase II clinical development that appear promising and are currently being explored. The field currently aims to determine the optimal sequence and combination of these therapies to best overcome such setbacks as toxicity and resistance and build upon the successes previously seen.
Collapse
Affiliation(s)
- Marlana Orloff
- Thomas Jefferson University, Department of Medical Oncology , 1015 Walnut Street, Philadelphia, PA 19107 , USA +1 215 955 1195 ; +1 215 923 0797 ;
| | | | | |
Collapse
|
1916
|
Subbiah V, McMahon C, Patel S, Zinner R, Silva EG, Elvin JA, Subbiah IM, Ohaji C, Ganeshan DM, Anand D, Levenback CF, Berry J, Brennan T, Chmielecki J, Chalmers ZR, Mayfield J, Miller VA, Stephens PJ, Ross JS, Ali SM. STUMP un"stumped": anti-tumor response to anaplastic lymphoma kinase (ALK) inhibitor based targeted therapy in uterine inflammatory myofibroblastic tumor with myxoid features harboring DCTN1-ALK fusion. J Hematol Oncol 2015; 8:66. [PMID: 26062823 PMCID: PMC4467062 DOI: 10.1186/s13045-015-0160-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 05/25/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Recurrent, metastatic mesenchymal myxoid tumors of the gynecologic tract present a management challenge as there is minimal evidence to guide systemic therapy. Such tumors also present a diagnostic dilemma, as myxoid features are observed in leiomyosarcomas, inflammatory myofibroblastic tumors (IMT), and mesenchymal myxoid tumors. Comprehensive genomic profiling was performed in the course of clinical care on a case of a recurrent, metastatic myxoid uterine malignancy (initially diagnosed as smooth muscle tumor of uncertain malignant potential (STUMP)), to guide identify targeted therapeutic options. To our knowledge, this case represents the first report of clinical response to targeted therapy in a tumor harboring a DCTN1-ALK fusion protein. METHODS Hybridization capture of 315 cancer-related genes plus introns from 28 genes often rearranged or altered in cancer was applied to >50 ng of DNA extracted from this sample and sequenced to high, uniform coverage. Therapy was given in the context of a phase I clinical trial ClinicalTrials.gov Identifier: ( NCT01548144 ). RESULTS Immunostains showed diffuse positivity for ALK1 expression and comprehensive genomic profiling identified an in frame DCTN1-ALK gene fusion. The diagnosis of STUMP was revised to that of an IMT with myxoid features. The patient was enrolled in a clinical trial and treated with an anaplastic lymphoma kinase (ALK) inhibitor (crizotinib/Xalkori®) and a multikinase VEGF inhibitor (pazopanib/Votrient®). The patient experienced an ongoing partial response (6+ months) by response evaluation criteria in solid tumors (RECIST) 1.1 criteria. CONCLUSIONS For myxoid tumors of the gynecologic tract, comprehensive genomic profiling can identify clinical relevant genomic alterations that both direct treatment targeted therapy and help discriminate between similar diagnostic entities.
Collapse
Affiliation(s)
- Vivek Subbiah
- Division of Cancer Medicine, Department of Investigational Cancer Therapeutic (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, FC8.3038, Box 0455, Houston, TX, 77030, USA.
| | | | - Shreyaskumar Patel
- Division of Cancer Medicine, Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Ralph Zinner
- Division of Cancer Medicine, Department of Investigational Cancer Therapeutic (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, FC8.3038, Box 0455, Houston, TX, 77030, USA.
| | - Elvio G Silva
- Division of Diagnostic Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Julia A Elvin
- Foundation Medicine, Inc, Cambridge, MA, 02141, USA.
| | - Ishwaria M Subbiah
- Division of Cancer Medicine, Department of Investigational Cancer Therapeutic (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, FC8.3038, Box 0455, Houston, TX, 77030, USA.
| | - Chimela Ohaji
- Division of Cancer Medicine, Department of Investigational Cancer Therapeutic (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, FC8.3038, Box 0455, Houston, TX, 77030, USA.
| | - Dhakshina Moorthy Ganeshan
- Division of Diagnostic Imaging and Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Deepa Anand
- Division of Diagnostic Imaging and Radiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Charles F Levenback
- Division of Surgery, Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Jenny Berry
- Division of Cancer Medicine, Department of Investigational Cancer Therapeutic (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, FC8.3038, Box 0455, Houston, TX, 77030, USA.
| | - Tim Brennan
- Foundation Medicine, Inc, Cambridge, MA, 02141, USA.
| | | | | | - John Mayfield
- Foundation Medicine, Inc, Cambridge, MA, 02141, USA.
| | | | | | | | - Siraj M Ali
- Foundation Medicine, Inc, Cambridge, MA, 02141, USA.
| |
Collapse
|
1917
|
Rissmann R, Hessel MHM, Cohen AF. Vemurafenib/dabrafenib and trametinib. Br J Clin Pharmacol 2015; 80:765-7. [PMID: 25847075 DOI: 10.1111/bcp.12651] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 03/27/2015] [Accepted: 03/30/2015] [Indexed: 01/20/2023] Open
Affiliation(s)
- Robert Rissmann
- Centre for Human Drug Research, Leiden, the Netherlands.,Leiden University Medical Center, Leiden, the Netherlands
| | | | - Adam F Cohen
- Centre for Human Drug Research, Leiden, the Netherlands.,Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
1918
|
BRAF inhibitors: the current and the future. Curr Opin Pharmacol 2015; 23:68-73. [PMID: 26072431 DOI: 10.1016/j.coph.2015.05.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 05/22/2015] [Accepted: 05/26/2015] [Indexed: 01/07/2023]
Abstract
The introduction of BRAF inhibitors (BRAFi), vemurafenib and dabrafenib, revolutionized BRAFV600-mutated metastatic melanoma treatment with improved response rate and overall survival compared to standard chemotherapy. However, the mechanism related cutaneous toxicity remains a concern. In addition, intrinsic and acquired resistance remain the key challenges in BRAFi therapy. Extensive efforts to elucidate the mechanisms have led to an improved understanding of disease biology and resulted in exploration of multiple new therapeutic options. While the future looks bright with multiple new therapeutic strategies in exploration and possible new generations of BRAFi, there are questions remaining to be answered to enable more efficient use of BRAFi in cancer therapy.
Collapse
|
1919
|
Deep Sequencing Reveals Occurrence of Subclonal ALK Mutations in Neuroblastoma at Diagnosis. Clin Cancer Res 2015; 21:4913-21. [DOI: 10.1158/1078-0432.ccr-15-0423] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/25/2015] [Indexed: 11/16/2022]
|
1920
|
Templeton IE, Musib L. MEK inhibitors beyond monotherapy: current and future development. Curr Opin Pharmacol 2015; 23:61-7. [PMID: 26057212 DOI: 10.1016/j.coph.2015.05.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 05/18/2015] [Accepted: 05/19/2015] [Indexed: 10/23/2022]
Abstract
The development of MEK inhibitors has led to improved progression-free survival in patients with mutant BRAF(V600) cancers when used in combination with BRAF inhibitors. However, resistance to combination therapy remains an issue. This review summarizes our current understanding of the role of MEK in cancer cell proliferation and the mechanisms which lead to resistance in patients. Specific adverse events, which have been linked to the MEK inhibitor class, have been described. Future combinations of MEK inhibitors with other cancer therapy options, currently under investigation in clinical trials, are also discussed.
Collapse
Affiliation(s)
- Ian E Templeton
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080 USA
| | - Luna Musib
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080 USA.
| |
Collapse
|
1921
|
Bol KF, Aarntzen EHJG, Hout FEMI', Schreibelt G, Creemers JHA, Lesterhuis WJ, Gerritsen WR, Grunhagen DJ, Verhoef C, Punt CJA, Bonenkamp JJ, de Wilt JHW, Figdor CG, de Vries IJM. Favorable overall survival in stage III melanoma patients after adjuvant dendritic cell vaccination. Oncoimmunology 2015; 5:e1057673. [PMID: 26942068 DOI: 10.1080/2162402x.2015.1057673] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 05/28/2015] [Accepted: 05/28/2015] [Indexed: 12/11/2022] Open
Abstract
Melanoma patients with regional metastatic disease are at high risk for recurrence and metastatic disease, despite radical lymph node dissection (RLND). We investigated the immunologic response and clinical outcome to adjuvant dendritic cell (DC) vaccination in melanoma patients with regional metastatic disease who underwent RLND with curative intent. In this retrospective study, 78 melanoma patients with regional lymph node metastasis who underwent RLND received autologous DCs loaded with gp100 and tyrosinase and were analyzed for functional tumor-specific T cell responses in skin-test infiltrating lymphocytes. The study shows that adjuvant DC vaccination in melanoma patients with regional lymph node metastasis is safe and induced functional tumor-specific T cell responses in 71% of the patients. The presence of functional tumor-specific T cells was correlated with a better 2-year overall survival (OS) rate. OS was significantly higher after adjuvant DC vaccination compared to 209 matched controls who underwent RLND without adjuvant DC vaccination, 63.6 mo vs. 31.0 mo (p = 0.018; hazard ratio 0.59; 95%CI 0.42-0.84). Five-year survival rate increased from 38% to 53% (p < 0.01). In summary, in melanoma patients with regional metastatic disease, who are at high risk for recurrence and metastatic disease after RLND, adjuvant DC vaccination is well tolerated. It induced functional tumor-specific immune responses in the majority of patients and these were related to clinical outcome. OS was significantly higher compared to matched controls. A randomized clinical trial is needed to prospectively validate the efficacy of DC vaccination in the adjuvant setting.
Collapse
Affiliation(s)
- Kalijn F Bol
- Department of Tumor Immunology; Radboud Institute for Molecular Life Sciences; Radboud University Medical Center; Nijmegen, The Netherlands; Department of Medical Oncology; Radboud University Medical Center; Nijmegen, The Netherlands
| | - Erik H J G Aarntzen
- Department of Tumor Immunology; Radboud Institute for Molecular Life Sciences; Radboud University Medical Center; Nijmegen, The Netherlands; Department of Medical Oncology; Radboud University Medical Center; Nijmegen, The Netherlands; Department of Radiology and Nuclear Medicine; Radboud University Medical Center; Nijmegen, The Netherlands
| | - Florentien E M In 't Hout
- Department of Tumor Immunology; Radboud Institute for Molecular Life Sciences; Radboud University Medical Center; Nijmegen, The Netherlands; Department of Surgical Oncology; Radboud University Medical Center; Nijmegen, The Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology; Radboud Institute for Molecular Life Sciences; Radboud University Medical Center ; Nijmegen, The Netherlands
| | - Jeroen H A Creemers
- Department of Tumor Immunology; Radboud Institute for Molecular Life Sciences; Radboud University Medical Center ; Nijmegen, The Netherlands
| | - W Joost Lesterhuis
- Department of Tumor Immunology; Radboud Institute for Molecular Life Sciences; Radboud University Medical Center; Nijmegen, The Netherlands; Department of Medicine and Pharmacology; University of Western Australia; Crawley, Australia
| | - Winald R Gerritsen
- Department of Medical Oncology; Radboud University Medical Center ; Nijmegen, The Netherlands
| | - Dirk J Grunhagen
- Department Surgical Oncology; Erasmus MC Cancer Institute ; Rotterdam, The Netherlands
| | - Cornelis Verhoef
- Department Surgical Oncology; Erasmus MC Cancer Institute ; Rotterdam, The Netherlands
| | - Cornelis J A Punt
- Department of Medical Oncology; Academic Medical Center ; Amsterdam, The Netherlands
| | - Johannes J Bonenkamp
- Department of Surgical Oncology; Radboud University Medical Center ; Nijmegen, The Netherlands
| | - Johannes H W de Wilt
- Department of Surgical Oncology; Radboud University Medical Center ; Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology; Radboud Institute for Molecular Life Sciences; Radboud University Medical Center ; Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Department of Tumor Immunology; Radboud Institute for Molecular Life Sciences; Radboud University Medical Center; Nijmegen, The Netherlands; Department of Medical Oncology; Radboud University Medical Center; Nijmegen, The Netherlands
| |
Collapse
|
1922
|
Rajakulendran T, Adam DN. Spotlight on pembrolizumab in the treatment of advanced melanoma. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:2883-6. [PMID: 26082618 PMCID: PMC4461129 DOI: 10.2147/dddt.s78036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Metastatic melanoma is an aggressive cancer with a poor prognosis. Many approved therapies often do not achieve durable responses in patients. This underscores the need for novel therapeutic strategies. Recruiting a robust immune response is an important antineoplastic treatment strategy. Immune checkpoints offer a molecular target for modulating the immune response and a promising therapeutic target in metastatic melanoma. Here we discuss the recent approval of pembrolizumab by the US Food and Drug Administration for the treatment of metastatic melanoma and its impact on future management of the disease.
Collapse
Affiliation(s)
- Thanashan Rajakulendran
- Department of Medicine, Division of Dermatology, Postgraduate Medical Education, University of Toronto, Toronto, ON, Canada
| | - David N Adam
- Department of Medicine, Division of Dermatology, St Michael's Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
1923
|
Sullivan RJ, Flaherty KT. New Strategies in Melanoma: Entering the Era of Combinatorial Therapy. Clin Cancer Res 2015; 21:2424-35. [DOI: 10.1158/1078-0432.ccr-14-1650] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
1924
|
Cherny NI, Sullivan R, Dafni U, Kerst JM, Sobrero A, Zielinski C, de Vries EGE, Piccart MJ. A standardised, generic, validated approach to stratify the magnitude of clinical benefit that can be anticipated from anti-cancer therapies: the European Society for Medical Oncology Magnitude of Clinical Benefit Scale (ESMO-MCBS). Ann Oncol 2015; 26:1547-73. [PMID: 26026162 DOI: 10.1093/annonc/mdv249] [Citation(s) in RCA: 621] [Impact Index Per Article: 62.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 05/22/2015] [Indexed: 12/12/2022] Open
Abstract
The value of any new therapeutic strategy or treatment is determined by the magnitude of its clinical benefit balanced against its cost. Evidence for clinical benefit from new treatment options is derived from clinical research, in particular phase III randomised trials, which generate unbiased data regarding the efficacy, benefit and safety of new therapeutic approaches. To date, there is no standard tool for grading the magnitude of clinical benefit of cancer therapies, which may range from trivial (median progression-free survival advantage of only a few weeks) to substantial (improved long-term survival). Indeed, in the absence of a standardised approach for grading the magnitude of clinical benefit, conclusions and recommendations derived from studies are often hotly disputed and very modest incremental advances have often been presented, discussed and promoted as major advances or 'breakthroughs'. Recognising the importance of presenting clear and unbiased statements regarding the magnitude of the clinical benefit from new therapeutic approaches derived from high-quality clinical trials, the European Society for Medical Oncology (ESMO) has developed a validated and reproducible tool to assess the magnitude of clinical benefit for cancer medicines, the ESMO Magnitude of Clinical Benefit Scale (ESMO-MCBS). This tool uses a rational, structured and consistent approach to derive a relative ranking of the magnitude of clinically meaningful benefit that can be expected from a new anti-cancer treatment. The ESMO-MCBS is an important first step to the critical public policy issue of value in cancer care, helping to frame the appropriate use of limited public and personal resources to deliver cost-effective and affordable cancer care. The ESMO-MCBS will be a dynamic tool and its criteria will be revised on a regular basis.
Collapse
Affiliation(s)
- N I Cherny
- Cancer Pain and Palliative Medicine Service, Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - R Sullivan
- Kings Health Partners Integrated Cancer Centre, King's College London, Institute of Cancer Policy, London, UK
| | - U Dafni
- University of Athens and Frontiers of Science Foundation-Hellas, Athens, Greece
| | - J M Kerst
- Department of Medical Oncology, Antoni van Leeuwenhoek Hospital
| | - A Sobrero
- Department of Medical Oncology, IRCCS San Martino IST, Genova, Italy
| | - C Zielinski
- Division of Oncology, Medical University Vienna, Vienna, Austria
| | - E G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - M J Piccart
- Jules Bordet Institute, UniversitéLibre de Bruxelles, Brussels, Belgium Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
1925
|
Massey PR, Prasad V, Figg WD, Fojo T. Multiplying therapies and reducing toxicity in metastatic melanoma. Cancer Biol Ther 2015; 16:1014-8. [PMID: 26016850 DOI: 10.1080/15384047.2015.1046650] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Prior to 2011, only 2 systemic treatments were approved for the treatment of melanoma and these had limited efficacy. In the past 4 years, 6 novel agents have received FDA approval. Herein, we will focus on 4 recently published NEJM papers reporting the results of clinical trials, comprising 4 agents targeting the MAPK pathway: the BRAF inhibitors vemurafenib and dabrafenib, and the MEK inhibitors trametinib and cobimetenib. These have been developed in parallel with a class of immunologic mediators often referred to as "immune checkpoint inhibitors." These recent studies represent a marked acceleration of progress in the treatment of metastatic melanoma. While it was hoped that combining BRAF and MEK inhibitors would significantly mitigate drug resistance, such combinations have yielded only modestly better results than monotherapy. However, these combinations were successful in reducing the development of cutaneous squamous cell carcinomas and keratocanthomas. Therefore, combination therapies are clearly warranted. Thus far there are only limited data addressing the value of combinations of immunotherapeutic agents: a phase 1 trial of concurrent nivolumab plus ipilimumab suggested enhanced activity that may not depend on BRAF mutation status. Despite the attention and publicity given to the progress achieved in the therapy of melanoma, the majority of patients with metastatic disease still have a poor prognosis. Even novel combination regiments of BRAF and MEK inhibitors achieve complete response in only 13% of patients and a median PFS of 11.4 months in all patients. Better therapies remain desperately needed, especially for the 30-40% of patients with wild-type BRAF, for whom BRAF/MAPK inhibition offers no benefit. In the latter benefit is expected from emerging immunotherapies either singly or in combinations. The extent to which immunotherapies will add to regimens targeting BRAF remains to be determined.
Collapse
Affiliation(s)
- Paul R Massey
- a Dell Medical School; The University of Texas at Austin ; Austin , TX , USA
| | | | | | | |
Collapse
|
1926
|
Postow MA, Chesney J, Pavlick AC, Robert C, Grossmann K, McDermott D, Linette GP, Meyer N, Giguere JK, Agarwala SS, Shaheen M, Ernstoff MS, Minor D, Salama AK, Taylor M, Ott PA, Rollin LM, Horak C, Gagnier P, Wolchok JD, Hodi FS. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med 2015; 372:2006-17. [PMID: 25891304 PMCID: PMC5744258 DOI: 10.1056/nejmoa1414428] [Citation(s) in RCA: 2208] [Impact Index Per Article: 220.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND In a phase 1 dose-escalation study, combined inhibition of T-cell checkpoint pathways by nivolumab and ipilimumab was associated with a high rate of objective response, including complete responses, among patients with advanced melanoma. METHODS In this double-blind study involving 142 patients with metastatic melanoma who had not previously received treatment, we randomly assigned patients in a 2:1 ratio to receive ipilimumab (3 mg per kilogram of body weight) combined with either nivolumab (1 mg per kilogram) or placebo once every 3 weeks for four doses, followed by nivolumab (3 mg per kilogram) or placebo every 2 weeks until the occurrence of disease progression or unacceptable toxic effects. The primary end point was the rate of investigator-assessed, confirmed objective response among patients with BRAF V600 wild-type tumors. RESULTS Among patients with BRAF wild-type tumors, the rate of confirmed objective response was 61% (44 of 72 patients) in the group that received both ipilimumab and nivolumab (combination group) versus 11% (4 of 37 patients) in the group that received ipilimumab and placebo (ipilimumab-monotherapy group) (P<0.001), with complete responses reported in 16 patients (22%) in the combination group and no patients in the ipilimumab-monotherapy group. The median duration of response was not reached in either group. The median progression-free survival was not reached with the combination therapy and was 4.4 months with ipilimumab monotherapy (hazard ratio associated with combination therapy as compared with ipilimumab monotherapy for disease progression or death, 0.40; 95% confidence interval, 0.23 to 0.68; P<0.001). Similar results for response rate and progression-free survival were observed in 33 patients with BRAF mutation-positive tumors. Drug-related adverse events of grade 3 or 4 were reported in 54% of the patients who received the combination therapy as compared with 24% of the patients who received ipilimumab monotherapy. Select adverse events with potential immunologic causes were consistent with those in a phase 1 study, and most of these events resolved with immune-modulating medication. CONCLUSIONS The objective-response rate and the progression-free survival among patients with advanced melanoma who had not previously received treatment were significantly greater with nivolumab combined with ipilimumab than with ipilimumab monotherapy. Combination therapy had an acceptable safety profile. (Funded by Bristol-Myers Squibb; ClinicalTrials.gov number, NCT01927419.).
Collapse
Affiliation(s)
- Michael A. Postow
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Jason Chesney
- J. Graham Brown Cancer Center, University of Louisville, Louisville, KY
| | | | - Caroline Robert
- Gustave, Roussy and Paris-Sud University, Villejuif-Paris-Sud, France
| | | | | | | | | | | | | | | | - Marc S. Ernstoff
- Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - David Minor
- California Pacific Center for Melanoma Research, San Francisco, CA
| | | | | | | | | | | | | | | | | |
Collapse
|
1927
|
Gibney GT, Gauthier G, Ayas C, Galebach P, Wu EQ, Abhyankar S, Reyes C, Guérin A, Yim YM. Treatment patterns and outcomes in BRAF V600E-mutant melanoma patients with brain metastases receiving vemurafenib in the real-world setting. Cancer Med 2015; 4:1205-13. [PMID: 25991583 PMCID: PMC4559032 DOI: 10.1002/cam4.475] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 04/15/2015] [Accepted: 04/21/2015] [Indexed: 01/22/2023] Open
Abstract
Brain metastases are a common and serious complication among patients with metastatic melanoma. The selective BRAF inhibitor vemurafenib has demonstrated clinical efficacy in patients with BRAF V600E-mutant melanoma brain metastases (MBM). We examined the real-world application and clinical outcomes of vemurafenib in this patient population. Demographic, treatment patterns, response, and survival data were collected from medical charts. Clinical data on 283 patients with active BRAF V600E-mutant MBM treated with vemurafenib were provided by 70 US oncologists. Mean age was 57.2 years, 60.8% were male, 67.5% had ECOG performance status of 0–1, and 43.1% used corticosteroids at vemurafenib initiation. Median follow-up was 5.7 months. Following vemurafenib initiation, 48.1% of patients experienced intracranial response and 45.6% experienced extracranial response. The Kaplan–Meier estimate for overall survival was 59% at 12 months. Multivariate analyses showed associations between intracranial response and both corticosteroid use and vemurafenib as initial therapy after MBM diagnosis. Larger size (5–10 mm vs. <5 mm) and number of brain metastases (≥5 vs. <2) and progressive extracranial disease at treatment initiation were associated with decreased intracranial response and increased risk of disease progression. Multiple extracranial sites (2 vs. <2) and the absence of local treatments were also associated with increased risk of progression. Increased risk of death was associated with ≥2 extracranial disease sites, progressive extracranial disease, and ≥5 brain metastases. Subgroups of MBM patients may derive more benefit with vemurafenib, warranting prospective investigation.
Collapse
Affiliation(s)
| | | | | | | | - Eric Q Wu
- Analysis Group Inc., Boston, Massachusetts
| | | | | | | | - Yeun Mi Yim
- Genentech Inc., South San Francisco, California
| |
Collapse
|
1928
|
Kohlhapp FJ, Broucek JR, Hughes T, Huelsmann EJ, Lusciks J, Zayas JP, Dolubizno H, Fleetwood VA, Grin A, Hill GE, Poshepny JL, Nabatiyan A, Ruby CE, Snook JD, Rudra JS, Schenkel JM, Masopust D, Zloza A, Kaufman HL. NK cells and CD8+ T cells cooperate to improve therapeutic responses in melanoma treated with interleukin-2 (IL-2) and CTLA-4 blockade. J Immunother Cancer 2015; 3:18. [PMID: 25992289 PMCID: PMC4437746 DOI: 10.1186/s40425-015-0063-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 04/17/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Melanoma is one of the few types of cancer with an increasing annual incidence. While a number of immunotherapies for melanoma have been associated with significant clinical benefit, including high-dose IL-2 and cytotoxic T lymphocyte antigen 4 (CTLA-4) blockade, clinical response to either of these single agents has been limited to 11-20% of treated patients. Therefore, in this study, we sought to test the hypothesis that the combination of IL-2 and CTLA-4 blockade could mediate a more profound therapeutic response. METHODS Here, B6 mice were challenged with poorly immunogenic B16 melanoma on day 0, and treated with CTLA-4 blocking antibody (100 μg/mouse) on days 3, 6, and 9, and IL-2 (100,000 units) twice daily on days 4-8, or both. RESULTS A highly significant synergistic effect that delayed tumor growth and prolonged survival was demonstrated with the combination immunotherapy compared to either monotherapy alone. The therapeutic effect of combination immunotherapy was dependent on both CD8+ T and NK cells and co-depletion of these subsets (but not either one alone) abrogated the therapeutic effect. CTLA-4 blockade increased immune cell infiltration (including CD8+ T cells and NK cells) in the tumor and IL-2 reduced the proportion of highly differentiated/exhausted tumor-infiltrating NK cells. CONCLUSIONS These results have implications for the design of clinical trials in patients with metastatic melanoma and provide new insights into how the immune system may be mediating anti-tumor activity with combination IL-2 and CTLA-4 blockade in melanoma.
Collapse
Affiliation(s)
- Frederick J Kohlhapp
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street Room 2007, New Brunswick, NJ 08901 USA.,Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612 USA
| | - Joseph R Broucek
- Department of General Surgery, Rush University Medical Center, Chicago, IL 60612 USA
| | - Tasha Hughes
- Department of General Surgery, Rush University Medical Center, Chicago, IL 60612 USA
| | - Erica J Huelsmann
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612 USA
| | - Jevgenijs Lusciks
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612 USA
| | - Janet P Zayas
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612 USA
| | - Hubert Dolubizno
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612 USA
| | | | - Alisa Grin
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612 USA
| | - Graham E Hill
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612 USA
| | - Joseph L Poshepny
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612 USA
| | - Arman Nabatiyan
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612 USA.,Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612 USA
| | - Carl E Ruby
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612 USA.,Department of General Surgery, Rush University Medical Center, Chicago, IL 60612 USA
| | - Joshua D Snook
- Department of Pharmacology & Toxicology and Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Jai S Rudra
- Department of Pharmacology & Toxicology and Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Jason M Schenkel
- Department of Microbiology and Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455 USA
| | - David Masopust
- Department of Microbiology and Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455 USA
| | - Andrew Zloza
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street Room 2007, New Brunswick, NJ 08901 USA.,Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL 60612 USA.,Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612 USA.,Department of Surgery, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903 USA
| | - Howard L Kaufman
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street Room 2007, New Brunswick, NJ 08901 USA.,Department of Surgery, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08903 USA
| |
Collapse
|
1929
|
Kass SL, Linden AF, Jackson PG, De Brito PA, Atkins MB. Bowel perforation associated with robust response to BRAF/MEK inhibitor therapy for BRAF-mutant melanoma: a case report. Melanoma Manag 2015; 2:115-120. [PMID: 30190840 DOI: 10.2217/mmt.15.10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Combination BRAF/MEK inhibition has shown improved response rates and longer progression-free and overall survival for patients with BRAF-mutant metastatic melanoma. A 63-year-old female with widely metastatic BRAF V600E-mutant melanoma was treated with dabrafenib/trametinib. Ten weeks into therapy, she was treated conservatively for a partial bowel obstruction involving a lesion in the distal ileum. She presented two weeks later with CT evidence of a high-grade bowel obstruction with perforation. Emergent surgery was performed. Intraoperative inspection and pathologic analysis of the resected specimen revealed no evidence of melanoma. Seven months postoperatively she is disease free and fully functional. Rapid BRAF/MEK inhibitor-induced regression of small bowel lesions can result in bowel perforation, which is critical to distinguish from the consequences of disease progression.
Collapse
Affiliation(s)
- Samantha L Kass
- Georgetown University School of Medicine, Washington, DC 20007, USA.,Georgetown University School of Medicine, Washington, DC 20007, USA
| | - Allison F Linden
- Department of Surgery, Georgetown University Medical Center, Washington, DC 20007, USA.,Department of Surgery, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Patrick G Jackson
- Department of Surgery, Georgetown University Medical Center, Washington, DC 20007, USA.,Department of Surgery, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Pedro A De Brito
- Department of Pathology, Georgetown University Medical Center, Washington, DC 20007, USA.,Department of Pathology, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Michael B Atkins
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC 20057, USA.,Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC 20057, USA
| |
Collapse
|
1930
|
Long-term outcome in BRAF(V600E) melanoma patients treated with vemurafenib: Patterns of disease progression and clinical management of limited progression. Eur J Cancer 2015; 51:1435-43. [PMID: 25980594 DOI: 10.1016/j.ejca.2015.04.010] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 04/10/2015] [Accepted: 04/16/2015] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Vemurafenib induces tumour regression in most patients with BRAF(V600E)-mutant melanoma; eventually, most experience progressive disease (PD). Long-term follow-up of patients with BRAF(V600E) melanoma treated in the phase 1 vemurafenib trial is reported. METHODS Patients received vemurafenib 240-1120 mg (dose escalation cohort) or 960 mg (extension cohort) orally twice daily. Clinical response was evaluated every 8 weeks by Response Evaluation Criteria In Solid Tumors (RECIST). Patients with PD amenable to local therapy (surgery or radiotherapy) were allowed to continue vemurafenib after progression. Overall survival (OS) from time of treatment initiation and from PD was estimated. Sites of PD were recorded. RESULTS Forty-eight patients (escalation cohort, n = 16; extension cohort, n = 32) received therapeutic doses of vemurafenib (⩾ 240 mg twice daily). Forty-four patients had PD by the time of this analysis and four remained progression free (follow-up time, 1.2-56.1 months). Median OS was 14 months (range, 1.2-56.1); 3- and 4-year melanoma-specific survival rate in the extension cohort was 26% and 19%, respectively. Median OS was 26.0 months (range, 7.7-56.1) among 20 patients who continued vemurafenib after local therapy. Median treatment duration beyond initial PD was 3.8 months (range, 1.1-26.6). In the extension cohort, six and five patients were alive after 3 and 4 years, respectively, on vemurafenib monotherapy. CONCLUSIONS Some patients with melanoma achieved long-term survival with vemurafenib monotherapy. Continuation of vemurafenib after PD might be beneficial in some patients because remaining disease might continue to respond to BRAF inhibition.
Collapse
|
1931
|
Kim DW, Barcena E, Mehta UN, Rohlfs ML, Kumar AJ, Penas-Prado M, Kim KB. Prolonged survival of a patient with metastatic leptomeningeal melanoma treated with BRAF inhibition-based therapy: a case report. BMC Cancer 2015; 15:400. [PMID: 25962795 PMCID: PMC4440312 DOI: 10.1186/s12885-015-1391-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Accepted: 04/29/2015] [Indexed: 02/05/2023] Open
Abstract
Background Leptomeningeal metastasis of melanoma is a devastating complication with a grave prognosis, and there are no known effective standard treatments. Although selective BRAF inhibitors have demonstrated a significant clinical activity in patients with metastatic melanoma harboring a BRAF mutation, the clinical benefit of BRAF inhibitor-based therapy in leptomeningeal disease is not clear. Case presentation We present a case of prolonged survival of a patient with BRAF V600E-mutant leptomeningeal disease who was treated with vemurafenib followed by whole brain radiation and a combination of dabrafenib and trametinib. Both vemurafenib and the sequential treatment of radiation and dabrafenib/trametinib led to regression of the leptomeningeal disease, and the patient survived for 19 months after the diagnosis of the leptomeningeal disease. Conclusion This case suggests a possible clinically meaningful benefit of BRAF inhibitor-based therapy and a need for close investigation of this therapeutic approach in patients with this devastating disease.
Collapse
Affiliation(s)
- Dae Won Kim
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Edelyn Barcena
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Urvi N Mehta
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Michelle L Rohlfs
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Ashok J Kumar
- Department of Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Marta Penas-Prado
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Kevin B Kim
- California Pacific Medical Center for Melanoma Research and Treatment, San Francisco Oncology Associates, 2333 Buchanan St., San Francisco, CA, 94115, USA.
| |
Collapse
|
1932
|
Gibney GT, Atkins MB. Swinging for the Fences: Long-Term Survival With Ipilimumab in Metastatic Melanoma. J Clin Oncol 2015; 33:1873-7. [PMID: 25964248 DOI: 10.1200/jco.2014.60.1807] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
A 40-year-old man with stage III melanoma arising from his left shoulder underwent wide local excision, sentinel lymph node biopsy, and lymph node dissection. Nine months after receiving adjuvant biochemotherapy with cisplatin, vinblastine, dacarbazine, interleukin-2 (IL-2), and interferon alfa as part of a clinical trial, he developed headaches and right-hand weakness and was found to have a 2-cm left parietal CNS metastasis. A comprehensive staging workup identified multiple nonspecific subcentimeter pulmonary nodules. The brain mass was resected and confirmed to be metastatic melanoma; the surgical bed was treated with stereotactic radiosurgery. He was monitored off therapy, but 5 months later, he developed a second left parietal CNS metastasis and enlarging lung nodules. The new brain lesion was treated with stereotactic radiosurgery, and he began systemic therapy with ipilimumab on a clinical trial. After the third dose, he presented with headache, nausea, and vomiting; a brain magnetic resonance imaging scan showed left anterior temporal enhancement, possibly representing new disease. His symptoms improved with a course of corticosteroids. Restaging of the chest showed a mixed response among the pulmonary nodules. After tapering off corticosteroids, he received the fourth dose of ipilimumab, which was complicated by grade 3 transaminitis and hypophysitis with documented hypothyroidism and adrenal insufficiency. They were managed with corticosteroids and thyroid and adrenal hormone replacement. Restaging scans showed further disease regression except for new confluent enhancing nodules and edema in the left temporal lobe. Craniotomy and resection of this area showed only necrotic tissue with no viable melanoma cells. Nine years after treatment with ipilimumab, he is alive and shows no evidence of melanoma on the basis of annual computed tomography scans of the chest, abdomen, and pelvis and magnetic resonance imaging scans of the brain. He has full neurologic function but still requires hormone replacement for persistent hypopituitarism.
Collapse
Affiliation(s)
| | - Michael B Atkins
- Georgetown-Lombardi Comprehensive Cancer Center; and Medstar-Georgetown University Hospital, Washington, DC
| |
Collapse
|
1933
|
Firor AE, Jares A, Ma Y. From humble beginnings to success in the clinic: Chimeric antigen receptor-modified T-cells and implications for immunotherapy. Exp Biol Med (Maywood) 2015; 240:1087-98. [PMID: 25956686 DOI: 10.1177/1535370215584936] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 03/31/2015] [Indexed: 11/15/2022] Open
Abstract
In the past 50 years, disease burden has steadily shifted from infectious disease to cancer. Standard chemotherapy has long been the mainstay of cancer medical management, and despite vast efforts towards more targeted and personalized drug therapy, many cancers remain refractory to treatment, with high rates of relapse and poor prognosis. Recent dramatic immunotherapy clinical trials have demonstrated that engineering T-cells with chimeric antigen receptors (CARs) to target CD19 can lead to complete remission in relapsed or refractory B-cell malignancies, generating a great deal of enthusiasm in the field. Here we provide a comprehensive overview of the history of adoptive T-cell therapy, including CARs, in solid tumors as well as hematologic malignancies. CAR therapy has the potential to fundamentally transform cancer treatment with specific and even personalized targeting of tissue- and tumor-specific antigens. However, before CARs become standard first-line treatment modalities, critical issues regarding efficacy, combinatorial regimens, and mechanisms of treatment failure and toxicity will need to be addressed.
Collapse
Affiliation(s)
- Amelia E Firor
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Alexander Jares
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA Medical Scientist Training Program, Stony Brook University, Stony Brook, NY 11794, USA Graduate Program in Molecular and Cellular Pharmacology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Yupo Ma
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
1934
|
Giannini I, Cutrignelli DA, Resta L, Gentile A, Vincenti L. Metastatic melanoma of the gallbladder: report of two cases and a review of the literature. Clin Exp Med 2015; 16:295-300. [PMID: 25929736 DOI: 10.1007/s10238-015-0353-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 04/20/2015] [Indexed: 12/14/2022]
Abstract
Melanoma is one of the most aggressive and highly metastatic cancers. The most common sites of distant metastases are soft tissues, lung, liver, skin and brain, whereas only few patients develop gastrointestinal metastases. Metastatic involvement of the gallbladder is rare and more often part of a widespread disease than a solitary lesion. The "gold-standard" treatment of metastatic melanoma of the gallbladder remains unclear. We report two cases of patients with past history of cutaneous melanoma who developed visceral metastases. The first patient was asymptomatic and had a widespread disease with metastatic involvement of both the spleen and the gallbladder. The second patient had an isolated metastasis of the gallbladder and complained of upper abdominal pain. The chosen treatment was open cholecystectomy (and splenectomy) in the first case and laparoscopic cholecystectomy in the second. A review of the literature is provided.
Collapse
Affiliation(s)
- I Giannini
- General Surgery and Liver Transplantation Unit, Department of Emergency and Organ Transplantation, University "A. Moro" of Bari, Policlinico. Piazza G. Cesare, 11, 70124, Bari, Italy.
| | - D A Cutrignelli
- Complex Operating Unit of Plastic Aesthetic and Reconstructive Surgery, Department of Emergency and Organ Transplantation, University "A. Moro" of Bari, Bari, Italy
| | - L Resta
- Department of Emergence and Organ Transplantation, Institute of Pathological Anatomy, University "A. Moro" of Bari, Bari, Italy
| | - A Gentile
- Department of Emergence and Organ Transplantation, Institute of Pathological Anatomy, University "A. Moro" of Bari, Bari, Italy
| | - L Vincenti
- General Surgery Unit, Policlinico of Bari, Bari, Italy
| |
Collapse
|
1935
|
|
1936
|
Gutzmer R. Aktueller Stand zu BRAF- und MEK-Inhibitoren in der Behandlung des malignen Melanoms. J Dtsch Dermatol Ges 2015; 13:485-7. [DOI: 10.1111/ddg.12692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
1937
|
Gross S, Rahal R, Stransky N, Lengauer C, Hoeflich KP. Targeting cancer with kinase inhibitors. J Clin Invest 2015; 125:1780-9. [PMID: 25932675 DOI: 10.1172/jci76094] [Citation(s) in RCA: 315] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Kinase inhibitors have played an increasingly prominent role in the treatment of cancer and other diseases. Currently, more than 25 oncology drugs that target kinases have been approved, and numerous additional therapeutics are in various stages of clinical evaluation. In this Review, we provide an in-depth analysis of activation mechanisms for kinases in cancer, highlight recent successes in drug discovery, and demonstrate the clinical impact of selective kinase inhibitors. We also describe the substantial progress that has been made in designing next-generation inhibitors to circumvent on-target resistance mechanisms, as well as ongoing strategies for combining kinase inhibitors in the clinic. Last, there are numerous prospects for the discovery of novel kinase targets, and we explore cancer immunotherapy as a new and promising research area for studying kinase biology.
Collapse
|
1938
|
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: 57] [Impact Index Per Article: 5.7] [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
|
1939
|
Abstract
Melanoma is a common cancer in the Western world with an increasing incidence. Sun exposure is still considered to be the major risk factor for melanoma. The prognosis of patients with malignant (advanced-stage) melanoma differs widely between countries, but public campaigns advocating early detection have led to significant reductions in mortality rates. As well as sun exposure, distinct genetic alterations have been identified as associated with melanoma. For example, families with melanoma who have germline mutations in CDKN2A are well known, whereas the vast majority of sporadic melanomas have mutations in the mitogen-activated protein kinase cascade, which is the pathway with the highest oncogenic and therapeutic relevance for this disease. BRAF and NRAS mutations are typically found in cutaneous melanomas, whereas KIT mutations are predominantly observed in mucosal and acral melanomas. GNAQ and GNA11 mutations prevail in uveal melanomas. Additionally, the PI3K-AKT-PTEN pathway and the immune checkpoint pathways are important. The finding that programmed cell death protein 1 ligand 1 (PDL1) and PDL2 are expressed by melanoma cells, T cells, B cells and natural killer cells led to the recent development of programmed cell death protein 1 (PD1)-specific antibodies (for example, nivolumab and pembrolizumab). Alongside other new drugs - namely, BRAF inhibitors (vemurafenib and dabrafenib) and MEK inhibitors (trametinib and cobimetinib) - these agents are very promising and have been shown to significantly improve prognosis for patients with advanced-stage metastatic disease. Early signs are apparent that these new treatment modalities are also improving long-term clinical benefit and the quality of life of patients. This Primer summarizes the current understanding of melanoma, from mechanistic insights to clinical progress. For an illustrated summary of this Primer, visit: http://go.nature.com/vX2N9s.
Collapse
|
1940
|
Sondermann W, Griewank KG, Schilling B, Livingstone E, Leyh JC, Rompoti N, Cosgarea I, Schimming T, Schadendorf D, Zimmer L, Hillen U. Corticosteroids augment BRAF inhibitor vemurafenib induced lymphopenia and risk of infection. PLoS One 2015; 10:e0124590. [PMID: 25897843 PMCID: PMC4405567 DOI: 10.1371/journal.pone.0124590] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 03/16/2015] [Indexed: 11/19/2022] Open
Abstract
We have previously demonstrated an impact of the BRAF inhibitor vemurafenib on patient lymphocyte counts. In the current study, the extent to which concomitant use of corticosteroids in BRAF inhibitor treated patients affects lymphocyte counts and predisposes to infection was investigated. A cohort of 102 patients receiving either the selective BRAF inhibitor vemurafenib or dabrafenib was analyzed. The amount of patients receiving either medication with or without systemic corticosteroids (dexamethasone) was determined and lymphocyte counts before and under therapy assessed. Additionally, the number and severity of infections occurring in these groups was analyzed. Vemurafenib treatment led to a considerable decrease in lymphocyte cell counts, with 62.3% of patients having lymphopenia. Dabrafenib treated patients only rarely demonstrated lymphopenia (12.5%). Dexamethasone co-administration further diminished lymphocyte counts. Lymphopenias were observed in 84.6% of patients receiving vemurafenib and dexamethasone. In our cohort, infections were noted in 9 patients, 4 of these were severe and 2 eventually fatal. All 9 cases with infections demonstrated lymphopenia, 8 of these had received dexamethasone and 7 of these a therapy with vemurafenib. Our findings demonstrate a significant lymphopenia in patients treated with the BRAF inhibitor vemurafenib, which is further augmented by dexamethasone and predisposes to infection. If validated in other studies, risk of infection should be considered when applying corticosteroids in combination with BRAF inhibitors, in particular vemurafenib.
Collapse
Affiliation(s)
- Wiebke Sondermann
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Klaus G. Griewank
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
- * E-mail: (KGG); (UH)
| | - Bastian Schilling
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Elisabeth Livingstone
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Julia C. Leyh
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Natalia Rompoti
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Ioana Cosgarea
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Tobias Schimming
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Uwe Hillen
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
- * E-mail: (KGG); (UH)
| |
Collapse
|
1941
|
Wattson DA, Sullivan RJ, Niemierko A, Merritt RM, Lawrence DP, Oh KS, Flaherty KT, Shih HA. Survival patterns following brain metastases for patients with melanoma in the MAP-kinase inhibitor era. J Neurooncol 2015; 123:75-84. [PMID: 25864098 DOI: 10.1007/s11060-015-1761-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 03/28/2015] [Indexed: 01/18/2023]
Abstract
Survival with BRAF-mutant metastatic melanoma is prolonged with MAP-kinase pathway inhibitors (MAPKi). Among patients with brain metastases (BM), however, the clinical course of MAPKi-treated patients is not well described. We therefore explored these patients' survival patterns compared to contemporary patients not treated with MAPKi. We analyzed 106 patients who developed melanoma BM between 2007 and 2013. Of these, 37 (35%) received de novo MAPKi for BRAF-mutant disease, which preceded BM in 49%. Immunotherapy was given to 54% of MAPKi-treated patients and 94% of those who did not receive MAPKi. We evaluated the potential influence of patient characteristics, systemic therapies, and BM-directed treatments on time to appearance of new BM and overall survival. With a median follow-up of 8.0 months after initial BM, MAPKi use was an independent predictor of prolonged survival after BM diagnosis (median 14.1 vs 7.0 months, P = 0.03, adjusted hazard ratio 0.39). This survival advantage was driven by the 16.6-month median survival of patients who initiated MAPKi after BM were diagnosed, versus 5.6 months if initiated prior to BM development (P = 0.03). Median survival from the onset of any systemic metastases was 22 months regardless of the timing of MAPKi relative to BM appearance. Time to in-brain progression was longer among patients whose MAPKi course was started after BM diagnosis, but MAPKi initiation prior to BM diagnosis was associated with longer time to intracranial involvement. These findings are consistent with potential MAPKi activity in intracranial melanoma.
Collapse
Affiliation(s)
- Daniel A Wattson
- Harvard Radiation Oncology Program, 75 Francis Street, ASB1, L2, Boston, MA, 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
1942
|
Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 2015. [PMID: 25860605 DOI: 10.1016/j.cell.2015.03.030.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2022]
Abstract
Research in two fronts has enabled the development of therapies that provide significant benefit to cancer patients. One area stems from a detailed knowledge of mutations that activate or inactivate signaling pathways that drive cancer development. This work triggered the development of targeted therapies that lead to clinical responses in the majority of patients bearing the targeted mutation, although responses are often of limited duration. In the second front are the advances in molecular immunology that unveiled the complexity of the mechanisms regulating cellular immune responses. These developments led to the successful targeting of immune checkpoints to unleash anti-tumor T cell responses, resulting in durable long-lasting responses but only in a fraction of patients. In this Review, we discuss the evolution of research in these two areas and propose that intercrossing them and increasing funding to guide research of combination of agents represent a path forward for the development of curative therapies for the majority of cancer patients.
Collapse
Affiliation(s)
- Padmanee Sharma
- Department of Immunology, MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - James P Allison
- Department of Immunology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
1943
|
Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 2015; 161:205-14. [PMID: 25860605 PMCID: PMC5905674 DOI: 10.1016/j.cell.2015.03.030] [Citation(s) in RCA: 1769] [Impact Index Per Article: 176.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Indexed: 12/16/2022]
Abstract
Research in two fronts has enabled the development of therapies that provide significant benefit to cancer patients. One area stems from a detailed knowledge of mutations that activate or inactivate signaling pathways that drive cancer development. This work triggered the development of targeted therapies that lead to clinical responses in the majority of patients bearing the targeted mutation, although responses are often of limited duration. In the second front are the advances in molecular immunology that unveiled the complexity of the mechanisms regulating cellular immune responses. These developments led to the successful targeting of immune checkpoints to unleash anti-tumor T cell responses, resulting in durable long-lasting responses but only in a fraction of patients. In this Review, we discuss the evolution of research in these two areas and propose that intercrossing them and increasing funding to guide research of combination of agents represent a path forward for the development of curative therapies for the majority of cancer patients.
Collapse
Affiliation(s)
- Padmanee Sharma
- Department of Immunology, MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - James P Allison
- Department of Immunology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
1944
|
Stuart SA, Houel S, Lee T, Wang N, Old WM, Ahn NG. A Phosphoproteomic Comparison of B-RAFV600E and MKK1/2 Inhibitors in Melanoma Cells. Mol Cell Proteomics 2015; 14:1599-615. [PMID: 25850435 DOI: 10.1074/mcp.m114.047233] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Indexed: 11/06/2022] Open
Abstract
Inhibitors of oncogenic B-RAF(V600E) and MKK1/2 have yielded remarkable responses in B-RAF(V600E)-positive melanoma patients. However, the efficacy of these inhibitors is limited by the inevitable onset of resistance. Despite the fact that these inhibitors target the same pathway, combination treatment with B-RAF(V600E) and MKK1/2 inhibitors has been shown to improve both response rates and progression-free survival in B-RAF(V600E) melanoma patients. To provide insight into the molecular nature of the combinatorial response, we used quantitative mass spectrometry to characterize the inhibitor-dependent phosphoproteome of human melanoma cells treated with the B-RAF(V600E) inhibitor PLX4032 (vemurafenib) or the MKK1/2 inhibitor AZD6244 (selumetinib). In three replicate experiments, we quantified changes at a total of 23,986 phosphosites on 4784 proteins. This included 1317 phosphosites that reproducibly decreased in response to at least one inhibitor. Phosphosites that responded to both inhibitors grouped into networks that included the nuclear pore complex, growth factor signaling, and transcriptional regulators. Although the majority of phosphosites were responsive to both inhibitors, we identified 16 sites that decreased only in response to PLX4032, suggesting rare instances where oncogenic B-RAF signaling occurs in an MKK1/2-independent manner. Only two phosphosites were identified that appeared to be uniquely responsive to AZD6244. When cells were treated with the combination of AZD6244 and PLX4032 at subsaturating concentrations (30 nm), responses at nearly all phosphosites were additive. We conclude that AZD6244 does not substantially widen the range of phosphosites inhibited by PLX4032 and that the benefit of the drug combination is best explained by their additive effects on suppressing ERK1/2 signaling. Comparison of our results to another recent ERK1/2 phosphoproteomics study revealed a surprising degree of variability in the sensitivity of phosphosites to MKK1/2 inhibitors in human cell lines, revealing unexpected cell specificity in the molecular responses to pathway activation.
Collapse
Affiliation(s)
| | | | - Thomas Lee
- From the ‡Department of Chemistry and Biochemistry
| | - Nan Wang
- From the ‡Department of Chemistry and Biochemistry
| | | | - Natalie G Ahn
- From the ‡Department of Chemistry and Biochemistry, §BioFrontiers Institute, University of Colorado, Boulder, Colorado 80309
| |
Collapse
|
1945
|
Vogel CJ, Smit MA, Maddalo G, Possik PA, Sparidans RW, van der Burg SH, Verdegaal EM, Heck AJR, Samatar AA, Beijnen JH, Altelaar AFM, Peeper DS. Cooperative induction of apoptosis in NRAS mutant melanoma by inhibition of MEK and ROCK. Pigment Cell Melanoma Res 2015; 28:307-17. [PMID: 25728708 DOI: 10.1111/pcmr.12364] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 02/25/2015] [Indexed: 12/13/2022]
Abstract
No effective targeted therapy is currently available for NRAS mutant melanoma. Experimental MEK inhibition is rather toxic and has only limited efficacy in clinical trials. At least in part, this is caused by the emergence of drug resistance, which is commonly seen for single agent treatment and shortens clinical responses. Therefore, there is a dire need to identify effective companion drug targets for NRAS mutant melanoma. Here, we show that at concentrations where single drugs had little effect, ROCK inhibitors GSK269962A or Fasudil, in combination with either MEK inhibitor GSK1120212 (Trametinib) or ERK inhibitor SCH772984 cooperatively caused proliferation inhibition and cell death in vitro. Simultaneous inhibition of MEK and ROCK caused induction of BimEL , PARP, and Puma, and hence apoptosis. In vivo, MEK and ROCK inhibition suppressed growth of established tumors. Our findings warrant clinical investigation of the effectiveness of combinatorial targeting of MAPK/ERK and ROCK in NRAS mutant melanoma.
Collapse
Affiliation(s)
- Celia J Vogel
- Division of Molecular Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1946
|
Abstract
INTRODUCTION Aberrant MAPK pathway signaling is a hallmark of melanoma. Mitogen/extracellular signal-regulated kinase (MEK) 1/2 are integral components of MAPK signaling. Several MEK inhibitors have demonstrated activity as single agents and in combination with other therapies. Trametinib was the first MEK inhibitor approved for use in treatment of advanced BRAF(V600) mutant melanoma as a single agent and in combination with BRAF inhibitor, dabrafenib. AREAS COVERED In this article, we discuss the underlying biology of MEK inhibition and its rationale in melanoma treatment with special emphasis on the clinical development of trametinib, from initial Phase I studies to randomized Phase II and III studies, both as monotherapy and in combination with other therapeutics. Furthermore, we briefly comment on trametinib for NRAS mutant and other non-BRAF mutant subsets of melanoma. EXPERT OPINION Trametinib is a novel oral MEK inhibitor with clinical activity in BRAF(V600) mutant metastatic melanoma alone and in combination with dabrafenib. Trametinib is currently being explored in other genetic subsets as well, particularly those with NRAS mutations or atypical BRAF alterations. Furthermore, to maximize efficacy and overcome acquired resistance, studies evaluating the combination of trametinib with other targeted agents and immunotherapy are underway.
Collapse
Affiliation(s)
- Ramya Thota
- Vanderbilt University Medical Center, Department of Medicine , 777 PRB, 2220 Pierce Ave, Nashville, TN 37232 , USA +1 615 322 8131 ; +1 615 343 7602 ;
| | | | | |
Collapse
|
1947
|
Heigener DF, Gandara DR, Reck M. Targeting of MEK in lung cancer therapeutics. THE LANCET RESPIRATORY MEDICINE 2015; 3:319-27. [PMID: 25801412 DOI: 10.1016/s2213-2600(15)00026-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The MAP-kinase pathway, consisting of the kinases RAS, RAF, MEK, and ERK, is crucial for cell proliferation, inhibition of apoptosis, and migration of cells. Direct inhibition of RAS is not yet possible, whereas inhibition of RAF is already established in malignant melanoma and under investigation in non-small-cell lung cancer (NSCLC). Due to their structure and function, the MEK proteins are attractive targets for cancer therapy and are also under investigation in NSCLC. We discuss strategies of targeting the RAS-RAF-MEK-ERK pathway with emphasis on MEK inhibition, either alone or in combination with other targets or conventional chemotherapy.
Collapse
Affiliation(s)
- David F Heigener
- Department of Thoracic Oncology, LungenClinic Grosshansdorf; member of the Airway research center north (ARCN) as part of the German Centre for Lung Research (DZL), Grosshansdorf, Germany.
| | - David R Gandara
- Division of Hematology/Oncology, UC Davis Comprehensive Cancer Center, Sacramento, USA
| | - Martin Reck
- Department of Thoracic Oncology, LungenClinic Grosshansdorf; member of the Airway research center north (ARCN) as part of the German Centre for Lung Research (DZL), Grosshansdorf, Germany
| |
Collapse
|
1948
|
Abstract
Hairy cell leukemia was initially described as a distinct entity in 1958. It is rare B-cell malignancy characterized by an indolent course. Advances in the treatment and understanding of the biology of hairy cell leukemia have made the disease exquisitely amenable to treatment. This review summarizes the present understanding of hairy cell leukemia with a particular focus on the development of novel and targeted approaches to treatment.
Collapse
|
1949
|
Schadendorf D, Amonkar MM, Stroyakovskiy D, Levchenko E, Gogas H, de Braud F, Grob JJ, Bondarenko I, Garbe C, Lebbe C, Larkin J, Chiarion-Sileni V, Millward M, Arance A, Mandalà M, Flaherty KT, Nathan P, Ribas A, Robert C, Casey M, DeMarini DJ, Irani JG, Aktan G, Long GV. Health-related quality of life impact in a randomised phase III study of the combination of dabrafenib and trametinib versus dabrafenib monotherapy in patients with BRAF V600 metastatic melanoma. Eur J Cancer 2015; 51:833-40. [PMID: 25794603 DOI: 10.1016/j.ejca.2015.03.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 03/04/2015] [Indexed: 01/23/2023]
Abstract
AIM To present the impact of treatments on health-related quality of life (HRQoL) from the double-blind, randomised phase III COMBI-d study that investigated the combination of dabrafenib and trametinib versus dabrafenib monotherapy in patients with BRAF V600E/K-mutant metastatic melanoma. COMBI-d showed significantly prolonged progression-free survival for the combination. METHODS HRQoL was evaluated using the European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire-C30, a generic cancer questionnaire (completed at baseline, during study treatment, at progression and post progression) assessing various dimensions (global health/QoL, functional status, and symptom impact). A mixed-model, repeated-measures analyses of covariance evaluated differences between arms. RESULTS Questionnaire completion rates were >95% at baseline, >85% to week 40 and >70% at disease progression. Baseline scores across both arms were comparable for all dimensions. Global health dimension scores were significantly better at weeks 8, 16 and 24 for patients receiving the combination during treatment and at progression. The majority of functional dimension scores (physical, social, role, emotional and cognitive functioning) trended in favour of the combination. Pain scores were significantly improved and clinically meaningful (6-13 point difference) for patients receiving the combination for all follow-up assessments versus those receiving dabrafenib monotherapy. For other symptom dimensions (nausea and vomiting, diarrhoea, dyspnoea, and constipation), scores trended in favour of dabrafenib monotherapy. CONCLUSION This analysis demonstrates that the combination of dabrafenib and trametinib provides better preservation of HRQoL and pain improvements versus dabrafenib monotherapy while also delaying progression. (Clinicaltrials.gov registration number: NCT01584648).
Collapse
Affiliation(s)
- Dirk Schadendorf
- Universitätsklinikum Essen, Hufelandstr. 55, Essen 45147, Germany.
| | - Mayur M Amonkar
- GlaxoSmithKline, 1250 S Collegeville Rd, Collegeville, PA 19426, United States.
| | | | - Evgeny Levchenko
- Petrov Research Institute of Oncology, 68 Leningradskaya Street, Saint Petersburg 197758, Russia.
| | - Helen Gogas
- University of Athens, Aghiou Thoma 17, Athens 11527, Greece.
| | - Filippo de Braud
- Fondazione IRCCS Istituto Nazionale Tumori, via Giacomo Venezian, 1, Milan, Italy.
| | - Jean-Jacques Grob
- Service de Dermatologie, Centre Hospitalo-Universitaire Sainte-Marguerite, 270 Boulevard de Sainte-Marguerite, Marseille 13009, France.
| | - Igor Bondarenko
- Dnepropetrovsk State Medical Academy, Dzerzhyns'koho Street 9, Dnepropetrovsk 49044, Ukraine.
| | - Claus Garbe
- Department of Dermatology, University Hospital Tuebingen, Liebermeisterstraße 25, Tuebingen 72076, Germany.
| | - Celeste Lebbe
- APHP Dermatology CIC Hôpital Saint Louis, University Paris Diderot, INSERM U976, Avenue Claude Vellefaux, Paris 75010, France.
| | - James Larkin
- Royal Marsden Hospital, Fulham Road, London SW3 6JJ, United Kingdom.
| | - Vanna Chiarion-Sileni
- Melanoma and Esophageal Oncology Unit, Veneto Oncology Institute-IRCCS, via Gattamelata, 64, Padua 35128, Italy.
| | - Michael Millward
- Sir Charles Gairdner Hospital, Hospital Avenue, Perth, WA 6009, Australia.
| | - Ana Arance
- Hospital Clinic, Carrer Villarroel 170, Barcelona 08036, Spain.
| | - Mario Mandalà
- Papa Giovanni XIII Hospital, Piazza OMS 1, Bergamo 24127, Italy.
| | - Keith T Flaherty
- Massachusetts General Hospital Cancer Center, 55 Fruit St, Boston 02114, MA, United States.
| | - Paul Nathan
- Mount Vernon Cancer Centre, Rickmansworth Road, Northwood HA6 2RN, United Kingdom.
| | - Antoni Ribas
- David Geffen School of Medicine, UCLA, 10833 Le Conte Avenue, Los Angeles 90095, CA, United States.
| | - Caroline Robert
- Gustave Roussy and Paris 11 University, 114 Rue Edouard Vaillant, Villejuif-Paris-Sud 94805, France.
| | - Michelle Casey
- GlaxoSmithKline, 1250 S Collegeville Rd, Collegeville, PA 19426, United States.
| | - Douglas J DeMarini
- GlaxoSmithKline, 1250 S Collegeville Rd, Collegeville, PA 19426, United States.
| | - Jhangir G Irani
- GlaxoSmithKline, 1250 S Collegeville Rd, Collegeville, PA 19426, United States.
| | - Gursel Aktan
- GlaxoSmithKline, 1250 S Collegeville Rd, Collegeville, PA 19426, United States.
| | - Georgina V Long
- Melanoma Institute Australia & The University of Sydney, 40 Rocklands Road, Sydney 2060, Australia.
| |
Collapse
|
1950
|
Hardy KM, Strizzi L, Margaryan NV, Gupta K, Murphy GF, Scolyer RA, Hendrix MJC. Targeting nodal in conjunction with dacarbazine induces synergistic anticancer effects in metastatic melanoma. Mol Cancer Res 2015; 13:670-80. [PMID: 25767211 DOI: 10.1158/1541-7786.mcr-14-0077] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 01/05/2015] [Indexed: 12/11/2022]
Abstract
UNLABELLED Metastatic melanoma is a highly aggressive skin cancer with a poor prognosis. Despite a complete response in fewer than 5% of patients, the chemotherapeutic agent dacarbazine (DTIC) remains the reference drug after almost 40 years. More recently, FDA-approved drugs have shown promise but patient outcome remains modest, predominantly due to drug resistance. As such, combinatorial targeting has received increased attention, and will advance with the identification of new molecular targets. One attractive target for improving melanoma therapy is the growth factor Nodal, whose normal expression is largely restricted to embryonic development, but is reactivated in metastatic melanoma. In this study, we sought to determine how Nodal-positive human melanoma cells respond to DTIC treatment and to ascertain whether targeting Nodal in combination with DTIC would be more effective than monotherapy. A single treatment with DTIC inhibited cell growth but did not induce apoptosis. Rather than reducing Nodal expression, DTIC increased the size of the Nodal-positive subpopulation, an observation coincident with increased cellular invasion. Importantly, clinical tissue specimens from patients with melanomas refractory to DTIC therapy stained positive for Nodal expression, both in pre- and post-DTIC tumors, underscoring the value of targeting Nodal. In vitro, anti-Nodal antibodies alone had some adverse effects on proliferation and apoptosis, but combining DTIC treatment with anti-Nodal antibodies decreased cell growth and increased apoptosis synergistically, at concentrations incapable of producing meaningful effects as monotherapy. IMPLICATIONS Targeting Nodal in combination with DTIC therapy holds promise for the treatment of metastatic melanoma.
Collapse
Affiliation(s)
- Katharine M Hardy
- Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Luigi Strizzi
- Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Naira V Margaryan
- Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Kanika Gupta
- Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois. Howard Hughes Medical Institute NU Bioscientist Program, Weinberg College of Arts and Sciences, Northwestern University, Evanston, Illinois
| | - George F Murphy
- Department of Pathology, Harvard Medical School, Brigham & Women's Hospital, Boston, Massachusetts
| | - Richard A Scolyer
- Melanoma Institute Australia; Sydney Medical School, The University of Sydney; and Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Mary J C Hendrix
- Program in Cancer Biology and Epigenomics, Stanley Manne Children's Research Institute at Ann and Robert H. Lurie Children's Hospital of Chicago, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|