101
|
Augustyn K, Joseph J, Patel AB, Razmandi A, Ali AN, Tawbi HA. Treatment experience with encorafenib plus binimetinib for BRAF V600-mutant metastatic melanoma: management insights for clinical practice. Melanoma Res 2023; 33:406-416. [PMID: 37534686 PMCID: PMC10470431 DOI: 10.1097/cmr.0000000000000891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 02/24/2023] [Indexed: 08/04/2023]
Abstract
For patients with locally advanced or metastatic melanoma who have BRAF V600 activating mutations, combination therapy with BRAF and MEK inhibitors is now the standard of care. The combination of encorafenib, a highly selective adenosine triphosphate-competitive BRAF inhibitor, plus binimetinib, a potent, selective, allosteric, non-adenosine triphosphate-competitive MEK1/2 inhibitor, was approved by the US Food and Drug Administration for unresectable or metastatic melanoma with BRAF V600E or V600K mutations based on data from the phase III COLUMBUS study (NCT01909453). Clinical data evaluating BRAF and MEK inhibitor combinations in advanced melanoma indicate a specific profile of adverse events that includes serious retinopathy, skin disorders, and cardiovascular toxicities. Here we provide an overview of the rationale for combining BRAF and MEK inhibitors for the treatment of melanoma, long-term safety results from COLUMBUS, and guidance on managing the most common adverse events associated with this combination based on clinical experience. Proactive and appropriate management of adverse events can allow for longer treatment durations and may result in better treatment outcomes.
Collapse
Affiliation(s)
- Kourtney Augustyn
- Department of Melanoma Medical Oncology, Division of Cancer Medicine
| | | | | | - Azadeh Razmandi
- Department of Head and Neck Surgery, Division of Ophthalmology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Amatul Noor Ali
- Department of Head and Neck Surgery, Division of Ophthalmology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hussein A. Tawbi
- Department of Melanoma Medical Oncology, Division of Cancer Medicine
| |
Collapse
|
102
|
Özdemir D, Büssgen M. Effectiveness and cost-effectiveness of combination therapy versus monotherapy in malignant melanoma. J Pharm Policy Pract 2023; 16:106. [PMID: 37749653 PMCID: PMC10521452 DOI: 10.1186/s40545-023-00611-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 09/16/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUND Until 2010, stage III or IV malignant melanoma (MM) had a poor prognosis. The discovery of immune checkpoint inhibitors (ICIs) in 2011 changed the treatment landscape. Promising results in patient survival with a checkpoint inhibitor prompted research into combination therapies. In 2016, the first combination therapy has been approved as first-line therapy for advanced MM. OBJECTIVE The aim of this work is to investigate to what extent combination therapy is (cost-)effective compared to monotherapy in stage III or IV MM. METHODS A systematic literature search was performed (Web of Science, PubMed, PubPharm, EconLit, and Cochrane Library); searching for publications published over the past decade that examine the cost-effectiveness in terms of cost/QALY and the effectiveness in terms of survival and response of combination therapy in comparison to monotherapy in stage III or IV MM patients. RESULTS A total of 11 randomized controlled trials (RCTs) and five cost-utility analyses met our inclusion criteria. Nine clinical trials demonstrated superiority of combination therapy over monotherapy. The combination of B-rapidly accelerated fibrosarcoma (BRAF) protein and mitogen-activated kinase (MEK) protein inhibitors is not cost-effective in any country. Three analyses demonstrate the cost-effectiveness of combination therapy with ICI compared to monotherapy. CONCLUSION Combination therapy is more effective compared to monotherapy. While combined ICIs are cost-effective compared to monotherapy, this is not the case for the combination of BRAF and MEK inhibitors.
Collapse
Affiliation(s)
| | - Melanie Büssgen
- Hamburg Center for Health Economics, University of Hamburg, Hamburg, Germany.
| |
Collapse
|
103
|
Bouffet E, Hansford JR, Garrè ML, Hara J, Plant-Fox A, Aerts I, Locatelli F, van der Lugt J, Papusha L, Sahm F, Tabori U, Cohen KJ, Packer RJ, Witt O, Sandalic L, Bento Pereira da Silva A, Russo M, Hargrave DR. Dabrafenib plus Trametinib in Pediatric Glioma with BRAF V600 Mutations. N Engl J Med 2023; 389:1108-1120. [PMID: 37733309 DOI: 10.1056/nejmoa2303815] [Citation(s) in RCA: 123] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
BACKGROUND Detection of the BRAF V600E mutation in pediatric low-grade glioma has been associated with a lower response to standard chemotherapy. In previous trials, dabrafenib (both as monotherapy and in combination with trametinib) has shown efficacy in recurrent pediatric low-grade glioma with BRAF V600 mutations, findings that warrant further evaluation of this combination as first-line therapy. METHODS In this phase 2 trial, patients with pediatric low-grade glioma with BRAF V600 mutations who were scheduled to receive first-line therapy were randomly assigned in a 2:1 ratio to receive dabrafenib plus trametinib or standard chemotherapy (carboplatin plus vincristine). The primary outcome was the independently assessed overall response (complete or partial response) according to the Response Assessment in Neuro-Oncology criteria. Also assessed were the clinical benefit (complete or partial response or stable disease for ≥24 weeks) and progression-free survival. RESULTS A total of 110 patients underwent randomization (73 to receive dabrafenib plus trametinib and 37 to receive standard chemotherapy). At a median follow-up of 18.9 months, an overall response occurred in 47% of the patients treated with dabrafenib plus trametinib and in 11% of those treated with chemotherapy (risk ratio, 4.31; 95% confidence interval [CI], 1.7 to 11.2; P<0.001). Clinical benefit was observed in 86% of the patients receiving dabrafenib plus trametinib and in 46% receiving chemotherapy (risk ratio, 1.88; 95% CI, 1.3 to 2.7). The median progression-free survival was significantly longer with dabrafenib plus trametinib than with chemotherapy (20.1 months vs. 7.4 months; hazard ratio, 0.31; 95% CI, 0.17 to 0.55; P<0.001). Grade 3 or higher adverse events occurred in 47% of the patients receiving dabrafenib plus trametinib and in 94% of those receiving chemotherapy. CONCLUSIONS Among pediatric patients with low-grade glioma with BRAF V600 mutations, dabrafenib plus trametinib resulted in significantly more responses, longer progression-free survival, and a better safety profile than standard chemotherapy as first-line therapy. (Funded by Novartis; ClinicalTrials.gov number, NCT02684058.).
Collapse
Affiliation(s)
- Eric Bouffet
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Jordan R Hansford
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Maria Luisa Garrè
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Junichi Hara
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Ashley Plant-Fox
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Isabelle Aerts
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Franco Locatelli
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Jasper van der Lugt
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Ludmila Papusha
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Felix Sahm
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Uri Tabori
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Kenneth J Cohen
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Roger J Packer
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Olaf Witt
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Larissa Sandalic
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Ana Bento Pereira da Silva
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Mark Russo
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| | - Darren R Hargrave
- From the Hospital for Sick Children, University of Toronto, Toronto (E.B., U.T.); the Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, VIC, and the Women's and Children's Hospital, South Australia Health and Medical Research Institute, South Australian immunoGENomics Cancer Institute, and the University of Adelaide, Adelaide - all in Australia (J.R.H.); IRCCS Giannina Gaslini Institute, Genoa (M.L.G.), and IRCCS Bambino Gesù Children's Hospital, Catholic University of the Sacred Heart, Rome (F.L.) - both in Italy; Osaka City General Hospital, Osaka, Japan (J.H.); the Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago (A.P.-F.); Institut Curie, SIREDO Oncology Center, Paris Sciences et Lettres Research University, Paris (I.A.); the Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands (J.L.); Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow (L.P.); the Department of Neuropathology and Clinical Cooperation Unit Neuropathology (F.S.) and the Hopp Children's Cancer Center, German Consortium for Translational Cancer Research, and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg University Hospital, Heidelberg, Germany (F.S., O.W.); the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore (K.J.C.); Children's National Hospital, Washington, D.C. (R.J.P.); Novartis Pharma, Basel, Switzerland (L.S., A.B.P.S.); Novartis Pharmaceuticals, East Hanover, NJ (M.R.); and the University College London Great Ormond Street Institute of Child Health, London (D.R.H.)
| |
Collapse
|
104
|
Myszkiewicz MF, Puzanov I, Goey AKL. Development and validation of an LC-MS/MS method to measure the BRAF inhibitors dabrafenib and encorafenib quantitatively and four major metabolites semi-quantitatively in human plasma. J Pharm Biomed Anal 2023; 234:115594. [PMID: 37478552 PMCID: PMC10528671 DOI: 10.1016/j.jpba.2023.115594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/28/2023] [Accepted: 07/16/2023] [Indexed: 07/23/2023]
Abstract
This article describes the development and validation of a liquid-chromatography coupled with tandem mass spectrometry (LC-MS/MS) assay for the simultaneous quantitation of the BRAF inhibitors dabrafenib and encorafenib, and semi-quantitation of their major metabolites (i.e., carboxy-dabrafenib, desmethyl-dabrafenib, hydroxy-dabrafenib, M42.5A) in human plasma. Analytes were extracted from human plasma by protein precipitation, followed by reversed phase high-performance liquid chromatography. Analyte detection was performed using tandem mass spectrometry with heated electrospray ionization operating in positive ion mode. The assay was validated in accordance with the current U.S. Food and Drug Administration Guidance on Bioanalytical Method Validation. Results showed that measurements were both accurate (94.6-112.0 %) and precise (within-run: 1.9-3.4 %; between-run: 1.7-12.0 %) spanning a concentration range of 5 to 2000 ng/mL for dabrafenib and 10 to 4000 ng/mL for encorafenib. Recoveries for these analytes were consistent with mean values ranging from 85.6 % to 90.9 %. The mean internal standard-normalized matrix factors for each drug ranged between 0.87 and 0.98 and were found to be precise (% RSD <6.4 %). Dabrafenib and encorafenib were stable in the final extract and in human plasma held under various storage conditions. The metabolites also passed the validation criteria for precision and selectivity. Finally, the clinical applicability of the assay was confirmed by (semi-)quantitation of all six analytes in plasma samples from cancer patients receiving standard-of-care treatment with dabrafenib and encorafenib. Reproducibility of the measured analyte concentrations in study samples was confirmed successfully by incurred sample reanalysis. In conclusion, this sensitive LC-MS/MS assay has been validated successfully and is suitable for therapeutic drug monitoring of dabrafenib and encorafenib and clinical pharmacokinetic studies with these BRAF inhibitors.
Collapse
Affiliation(s)
- Melody F Myszkiewicz
- Bioanalytics, Metabolomics, and Pharmacokinetics Shared Resource, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Andrew K L Goey
- Bioanalytics, Metabolomics, and Pharmacokinetics Shared Resource, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA; Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA.
| |
Collapse
|
105
|
Mezi S, Botticelli A, Scagnoli S, Pomati G, Fiscon G, De Galitiis F, Di Pietro FR, Verkhovskaia S, Amirhassankhani S, Pisegna S, Gentile G, Simmaco M, Gohlke B, Preissner R, Marchetti P. The Impact of Drug-Drug Interactions on the Toxicity Profile of Combined Treatment with BRAF and MEK Inhibitors in Patients with BRAF-Mutated Metastatic Melanoma. Cancers (Basel) 2023; 15:4587. [PMID: 37760556 PMCID: PMC10526382 DOI: 10.3390/cancers15184587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND BRAF and MEK inhibition is a successful strategy in managing BRAF-mutant melanoma, even if the treatment-related toxicity is substantial. We analyzed the role of drug-drug interactions (DDI) on the toxicity profile of anti-BRAF/anti-MEK therapy. METHODS In this multicenter, observational, and retrospective study, DDIs were assessed using Drug-PIN software (V 2/23). The association between the Drug-PIN continuous score or the Drug-PIN traffic light and the occurrence of treatment-related toxicities and oncological outcomes was evaluated. RESULTS In total, 177 patients with advanced BRAF-mutated melanoma undergoing BRAF/MEK targeted therapy were included. All grade toxicity was registered in 79% of patients. Cardiovascular toxicities occurred in 31 patients (17.5%). Further, 94 (55.9%) patients had comorbidities requiring specific pharmacological treatments. The median Drug-PIN score significantly increased when the target combination was added to the patient's home therapy (p-value < 0.0001). Cardiovascular toxicity was significantly associated with the Drug-PIN score (p-value = 0.048). The Drug-PIN traffic light (p = 0.00821) and the Drug-PIN score (p = 0.0291) were seen to be significant predictors of cardiotoxicity. Patients with low-grade vs. high-grade interactions showed a better prognosis regarding overall survival (OS) (p = 0.0045) and progression-free survival (PFS) (p = 0.012). The survival analysis of the subgroup of patients with cardiological toxicity demonstrated that patients with low-grade vs. high-grade DDIs had better outcomes in terms of OS (p = 0.0012) and a trend toward significance in PFS (p = 0.068). CONCLUSIONS DDIs emerged as a critical issue for the risk of treatment-related cardiovascular toxicity. Our findings support the utility of DDI assessment in melanoma patients treated with BRAF/MEK inhibitors.
Collapse
Affiliation(s)
- Silvia Mezi
- Department of Radiological, Oncological, and Anatomopathological Sciences, Sapienza University of Rome, 00161 Rome, Italy; (S.M.); (A.B.)
| | - Andrea Botticelli
- Department of Radiological, Oncological, and Anatomopathological Sciences, Sapienza University of Rome, 00161 Rome, Italy; (S.M.); (A.B.)
| | - Simone Scagnoli
- Department of Radiological, Oncological, and Anatomopathological Sciences, Sapienza University of Rome, 00161 Rome, Italy; (S.M.); (A.B.)
| | - Giulia Pomati
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.P.); (S.P.)
| | - Giulia Fiscon
- Department of Computer, Control, and Management Engineering “Antonio Ruberti”, Sapienza University of Rome, 00161 Rome, Italy;
| | - Federica De Galitiis
- Istituto Dermopatico dell’Immacolata, IDI-IRCCS, 00144 Rome, Italy; (F.D.G.); (F.R.D.P.); (S.V.); (P.M.)
| | - Francesca Romana Di Pietro
- Istituto Dermopatico dell’Immacolata, IDI-IRCCS, 00144 Rome, Italy; (F.D.G.); (F.R.D.P.); (S.V.); (P.M.)
| | - Sofia Verkhovskaia
- Istituto Dermopatico dell’Immacolata, IDI-IRCCS, 00144 Rome, Italy; (F.D.G.); (F.R.D.P.); (S.V.); (P.M.)
| | - Sasan Amirhassankhani
- Department of Urology, S. Orsola-Malpighi Hospital, University of Bologna, Via Palagi, 40126 Bologna, Italy;
| | - Simona Pisegna
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.P.); (S.P.)
| | - Giovanna Gentile
- Department of Neuroscience, Mental Health, and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University, 00185 Rome, Italy; (G.G.); (M.S.)
- Unit of Laboratory and Advanced Molecular Diagnostics, ‘Sant’Andrea’ University Hospital, 00189 Rome, Italy
| | - Maurizio Simmaco
- Department of Neuroscience, Mental Health, and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University, 00185 Rome, Italy; (G.G.); (M.S.)
- Unit of Laboratory and Advanced Molecular Diagnostics, ‘Sant’Andrea’ University Hospital, 00189 Rome, Italy
| | - Bjoern Gohlke
- Structural Bioinformatics Group, Institute for Physiology, Charité-University Medicine Berlin, 10117 Berlin, Germany; (B.G.); (R.P.)
| | - Robert Preissner
- Structural Bioinformatics Group, Institute for Physiology, Charité-University Medicine Berlin, 10117 Berlin, Germany; (B.G.); (R.P.)
| | - Paolo Marchetti
- Istituto Dermopatico dell’Immacolata, IDI-IRCCS, 00144 Rome, Italy; (F.D.G.); (F.R.D.P.); (S.V.); (P.M.)
| |
Collapse
|
106
|
Valery M, Vasseur D, Fachinetti F, Boilève A, Smolenschi C, Tarabay A, Antoun L, Perret A, Fuerea A, Pudlarz T, Boige V, Hollebecque A, Ducreux M. Targetable Molecular Alterations in the Treatment of Biliary Tract Cancers: An Overview of the Available Treatments. Cancers (Basel) 2023; 15:4446. [PMID: 37760415 PMCID: PMC10526255 DOI: 10.3390/cancers15184446] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/27/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Biliary tract cancers (BTCs) are rare tumours, most often diagnosed at an unresectable stage, associated with poor prognosis, with a 5-year survival rate not exceeding 10%. Only first- and second-line treatments are well codified with the combination of cisplatin-gemcitabine chemotherapy and immunotherapy followed by 5-FU and oxaliplatin chemotherapy, respectively. Many studies have shown that BTC, and more particularly intrahepatic cholangiocarcinoma (iCCA), have a high rate of targetable somatic alteration. To date, the FDA has approved several drugs. Ivosidenib targeting IDH1 mutations, as well as futibatinib and pemigatinib targeting FGFR2 fusions, are approved for pre-treated advanced CCA. The combination of dabrafenib and trametinib are approved for BRAFV600E mutated advanced tumours, NTRK inhibitors entrectinib and larotrectinib for tumours bearing NTRK fusion and prembrolizumab for MSI-H advanced tumours, involving a small percentage of BTC in these three settings. Several other potentially targetable alterations are found in BTC, such as HER2 mutations or amplifications or KRASG12C mutations and mutations in genes involved in DNA repair mechanisms. This review aims to clarify the specific diagnostic modalities for gene alterations and to summarize the results of the main trials and developments underway for the management of advanced BTC with targetable alterations.
Collapse
Affiliation(s)
- Marine Valery
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
| | - Damien Vasseur
- Medical Biology and Pathology Department, Gustave Roussy, F-94805 Villejuif, France;
| | - Francesco Fachinetti
- Dana-Farber Institute, Lowe Center for Thoracic Oncology, Boston, MA 02215, USA;
| | - Alice Boilève
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
- Université Paris-Saclay, Gustave Roussy, Inserm Unité Dynamique des Cellules Tumorales, F-94805 Villejuif, France
| | - Cristina Smolenschi
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
- Département d’Innovation Thérapeutique, Gustave Roussy, F-94805 Villejuif, France
| | - Anthony Tarabay
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
| | - Leony Antoun
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
| | - Audrey Perret
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
| | - Alina Fuerea
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
| | - Thomas Pudlarz
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
| | - Valérie Boige
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
| | - Antoine Hollebecque
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
- Département d’Innovation Thérapeutique, Gustave Roussy, F-94805 Villejuif, France
| | - Michel Ducreux
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
- Université Paris-Saclay, Gustave Roussy, Inserm Unité Dynamique des Cellules Tumorales, F-94805 Villejuif, France
| |
Collapse
|
107
|
Grossauer A, Uranowska K, Kitzwögerer M, Mostegel M, Breiteneder H, Hafner C. Immunohistochemical detection of the chondroitin sulfate proteoglycan 4 protein in primary and metastatic melanoma. Oncol Lett 2023; 26:382. [PMID: 37559576 PMCID: PMC10407859 DOI: 10.3892/ol.2023.13968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/22/2023] [Indexed: 08/11/2023] Open
Abstract
Treatment of malignant melanoma, the most aggressive form of skin cancer, continues to be a major challenge for clinicians. New targeted therapies with kinase inhibitors or drugs which modify the immune response are often accompanied by the development of resistance or severe side effects. In this context, chondroitin sulfate proteoglycan 4 (CSPG4), a highly immunogenic melanoma tumor antigen, could be a potential target for alternative therapeutic approaches. The aim of the present study was to identify differences in the levels of CSPG4 protein expression in primary and metastatic melanomas as well as to analyze correlations between CSPG4 expression and histopathological data and patient characteristics. A total of 189 melanoma tissue samples from Lower Austria, including primary melanomas and melanoma metastases, were immunohistochemically stained for the expression of CSPG4 and statistical analyses were performed. A total of 65.6% of melanoma tissue samples stained positive for the expression of CSPG4. Primary nodular and primary superficial spreading melanomas demonstrated a significantly higher number of positively stained tissue samples for CSPG4 compared with primary lentigo maligna melanomas. No significant differences in the expression of CSPG4 were demonstrated between primary melanomas and melanoma metastases. The present study supports the advancement of the understanding of CSPG4 tissue expression patterns in melanoma patients and provides additional information for further investigation of CSPG4 as a potential therapeutic target.
Collapse
Affiliation(s)
- Anna Grossauer
- Department of Dermatology, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, A-3100 St. Poelten, Austria
- Department of Pathology, University Hospital Krems, Karl Landsteiner University of Health Sciences, A-3500 Krems an der Donau, Austria
| | - Karolina Uranowska
- Department of Dermatology, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, A-3100 St. Poelten, Austria
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Melitta Kitzwögerer
- Department of Pathology, University Hospital St. Poelten, A-3100 St. Poelten, Austria
| | - Margit Mostegel
- Department of Pathology, University Hospital Krems, Karl Landsteiner University of Health Sciences, A-3500 Krems an der Donau, Austria
| | - Heimo Breiteneder
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Christine Hafner
- Department of Dermatology, University Hospital St. Poelten, Karl Landsteiner University of Health Sciences, A-3100 St. Poelten, Austria
- Karl Landsteiner Institute of Dermatological Research, Karl Landsteiner Gesellschaft, A-3100 St. Poelten, Austria
| |
Collapse
|
108
|
Fay CJ, Jakuboski S, Mclellan B, Allais BS, Semenov Y, Larocca CA, LeBoeuf NR. Diagnosis and Management of Dermatologic Adverse Events from Systemic Melanoma Therapies. Am J Clin Dermatol 2023; 24:765-785. [PMID: 37395930 PMCID: PMC10796164 DOI: 10.1007/s40257-023-00790-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2023] [Indexed: 07/04/2023]
Abstract
The advent of protein kinase inhibitors and immunotherapy has profoundly improved the management of advanced melanoma. However, with these therapeutic advancements also come drug-related toxicities that have the potential to affect various organ systems. We review dermatologic adverse events from targeted (including BRAF and MEK inhibitor-related) and less commonly used melanoma treatments, with a focus on diagnosis and management. As immunotherapy-related toxicities have been extensively reviewed, herein, we discuss injectable talimogene laherparepvec and touch on recent breakthroughs in the immunotherapy space. Dermatologic adverse events may severely impact quality of life and are associated with response and survival. It is therefore essential that clinicians are aware of their diverse presentations and management strategies.
Collapse
Affiliation(s)
- Christopher J Fay
- Department of Dermatology, Brigham and Women's Hospital, and the Center for Cutaneous Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, MA, USA
| | | | - Beth Mclellan
- Department of Dermatology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Blair S Allais
- Department of Dermatology, Brigham and Women's Hospital, and the Center for Cutaneous Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Yevgeniy Semenov
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cecilia A Larocca
- Department of Dermatology, Brigham and Women's Hospital, and the Center for Cutaneous Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Nicole R LeBoeuf
- Department of Dermatology, Brigham and Women's Hospital, and the Center for Cutaneous Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
109
|
Yamamoto S, Kuriyama H, Kashiwada-Nakamura K, Kajihara I, Makino K, Aoi J, Miyashita A, Yoshida C, Kubo Y, Fukushima S. A case of unknown primary melanoma detected with a BRAF gene mutation from a pleural fluid cell block. J Dermatol 2023; 50:e305-e306. [PMID: 37088959 DOI: 10.1111/1346-8138.16812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/04/2023] [Accepted: 04/10/2023] [Indexed: 04/25/2023]
Affiliation(s)
- Sotaro Yamamoto
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Haruka Kuriyama
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kayo Kashiwada-Nakamura
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Ikko Kajihara
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Katsunari Makino
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Jun Aoi
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Azusa Miyashita
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Chieko Yoshida
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yosuke Kubo
- Department of Dermatology, Kumamoto Red Cross Hospital, Kumamoto, Japan
| | - Satoshi Fukushima
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
110
|
van Akkooi AC, Hauschild A, Long GV, Mandala M, Kicinski M, Govaerts AS, Klauck I, Ouali M, Lorigan PC, Eggermont AM. COLUMBUS-AD: phase III study of adjuvant encorafenib + binimetinib in resected stage IIB/IIC BRAF V600-mutated melanoma. Future Oncol 2023; 19:2017-2027. [PMID: 37665297 DOI: 10.2217/fon-2023-0414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023] Open
Abstract
Stage IIB/IIC melanoma has a high risk of recurrence after surgical resection. While, for decades, surgery was the only option for high-risk stage II disease in most countries, adjuvant therapies now exist. Anti-programmed cell death protein 1 (PD-1) antibodies significantly improve recurrence-free survival versus placebo in patients with fully resected stage IIB/IIC melanoma. Combined BRAF MEK inhibitor therapy showed benefits in high-risk stage III and advanced disease; however, its role in patients with fully resected stage BRAF-mutated IIB/IIC melanoma is still unknown. Here we describe the rationale and design of the ongoing randomized, placebo-controlled COLUMBUS-AD trial, the first study of a BRAF-MEK inhibitor combination therapy (encorafenib + binimetinib) in patients with BRAF V600-mutated stage IIB/IIC melanoma.
Collapse
Affiliation(s)
- Alexander Cj van Akkooi
- Melanoma Institute Australia, the University of Sydney & Royal Prince Alfred Hospital, 40 Rocklands Road Wollstonecraft, Sydney 2065, NSW, Australia
| | - Axel Hauschild
- Department of Dermatology, University Hospital (UKSH), Arnold-Heller-Straße 3, 24105, Kiel, Germany
| | - Georgina V Long
- Melanoma Institute Australia, University of Sydney, & Mater & Royal North Shore Hospitals, 40 Rocklands Road Wollstonecraft, Sydney 2065, NSW, Australia
| | - Mario Mandala
- University of Perugia, Ospedale Santa Maria della Misericordia, Piazzale Giorgio Menghini, 3, 06129, Perugia, Italy
| | - Michal Kicinski
- EORTC Headquarters, Avenue Emmanuel Mounier 83/11, 1200, Brussels, Belgium
| | | | - Isabelle Klauck
- Pierre Fabre, Medical & Patient/Consumer Division, 33 avenue Emile Zola, 92100, Boulogne-Billancourt, France
| | - Monia Ouali
- Pierre Fabre, Medical & Patient/Consumer Division, Langlade, France
| | - Paul C Lorigan
- Christie NHS Foundation Trust, Wilmslow Road Manchester M20 4BX, UK
| | - Alexander Mm Eggermont
- University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
- Comprehensive Cancer Center Munich, Technical University Munich & Ludwig Maximiliaan University, Marchioninistraße 15, 81377 Munich, Germany
| |
Collapse
|
111
|
Qin Z, Zheng M. Advances in targeted therapy and immunotherapy for melanoma (Review). Exp Ther Med 2023; 26:416. [PMID: 37559935 PMCID: PMC10407994 DOI: 10.3892/etm.2023.12115] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 06/28/2023] [Indexed: 08/11/2023] Open
Abstract
Melanoma is the most aggressive and deadly type of skin cancer and is known for its poor prognosis as soon as metastasis occurs. Since 2011, new and effective therapies for metastatic melanoma have emerged, with US Food and Drug Administration approval of multiple targeted agents, such as V-Raf murine sarcoma viral oncogene homolog B1/mitogen-activated protein kinase kinase inhibitors and multiple immunotherapy agents, such as cytotoxic T lymphocyte-associated protein 4 and anti-programmed cell death protein 1/ligand 1 blockade. Based on insight into the respective advantages of the above two strategies, the present article provided a review of clinical trials of the application of targeted therapy and immunotherapy, as well as novel approaches of their combinations for the treatment of metastatic melanoma in recent years, with a focus on upcoming initiatives to improve the efficacy of these treatment approaches for metastatic melanoma.
Collapse
Affiliation(s)
- Ziyao Qin
- No. 4 Research Laboratory, Shanghai Institute of Biological Products Co., Ltd., Shanghai 200051, P.R. China
| | - Mei Zheng
- No. 4 Research Laboratory, Shanghai Institute of Biological Products Co., Ltd., Shanghai 200051, P.R. China
| |
Collapse
|
112
|
Budiman A, Handini AL, Muslimah MN, Nurani NV, Laelasari E, Kurniawansyah IS, Aulifa DL. Amorphous Solid Dispersion as Drug Delivery Vehicles in Cancer. Polymers (Basel) 2023; 15:3380. [PMID: 37631436 PMCID: PMC10457821 DOI: 10.3390/polym15163380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Cancer treatment has improved over the past decades, but a major challenge lies in drug formulation, specifically for oral administration. Most anticancer drugs have poor water solubility which can affect their bioavailability. This causes suboptimal pharmacokinetic performance, resulting in limited efficacy and safety when administered orally. As a result, it is essential to develop a strategy to modify the solubility of anticancer drugs in oral formulations to improve their efficacy and safety. A promising approach that can be implemented is amorphous solid dispersion (ASD) which can enhance the aqueous solubility and bioavailability of poorly water-soluble drugs. The addition of a polymer can cause stability in the formulations and maintain a high supersaturation in bulk medium. Therefore, this study aimed to summarize and elucidate the mechanisms and impact of an amorphous solid dispersion system on cancer therapy. To gather relevant information, a comprehensive search was conducted using keywords such as "anticancer drug" and "amorphous solid dispersion" in the PubMed, Scopus, and Google Scholar databases. The review provides an overview and discussion of the issues related to the ASD system used to improve the bioavailability of anticancer drugs based on molecular pharmaceutics. A thorough understanding of anticancer drugs in this system at a molecular level is imperative for the rational design of the products.
Collapse
Affiliation(s)
- Arif Budiman
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Annisa Luthfiyah Handini
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Mutia Nur Muslimah
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Neng Vera Nurani
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Eli Laelasari
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Insan Sunan Kurniawansyah
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia; (A.L.H.); (M.N.M.); (N.V.N.); (E.L.); (I.S.K.)
| | - Diah Lia Aulifa
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jl. Raya Bandung-Sumedang Km. 21, Bandung 45363, Indonesia;
| |
Collapse
|
113
|
Zhou JB, Tang D, He L, Lin S, Lei JH, Sun H, Xu X, Deng CX. Machine learning model for anti-cancer drug combinations: Analysis, prediction, and validation. Pharmacol Res 2023; 194:106830. [PMID: 37343647 DOI: 10.1016/j.phrs.2023.106830] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/10/2023] [Accepted: 06/17/2023] [Indexed: 06/23/2023]
Abstract
Drug combination therapy is a highly effective approach for enhancing the therapeutic efficacy of anti-cancer drugs and overcoming drug resistance. However, the innumerable possible drug combinations make it impractical to screen all synergistic drug pairs. Moreover, biological insights into synergistic drug pairs are still lacking. To address this challenge, we systematically analyzed drug combination datasets curated from multiple databases to identify drug pairs more likely to show synergy. We classified drug pairs based on their MoA and discovered that 110 MoA pairs were significantly enriched in synergy in at least one type of cancer. To improve the accuracy of predicting synergistic effects of drug pairs, we developed a suite of machine learning models that achieve better predictive performance. Unlike most previous methods that were rarely validated by wet-lab experiments, our models were validated using two-dimensional cell lines and three-dimensional tumor slice culture (3D-TSC) models, implying their practical utility. Our prediction and validation results indicated that the combination of the RTK inhibitors Lapatinib and Pazopanib exhibited a strong therapeutic effect in breast cancer by blocking the downstream PI3K/AKT/mTOR signaling pathway. Furthermore, we incorporated molecular features to identify potential biomarkers for synergistic drug pairs, and almost all potential biomarkers found connections between drug targets and corresponding molecular features using protein-protein interaction network. Overall, this study provides valuable insights to complement and guide rational efforts to develop drug combination treatments.
Collapse
Affiliation(s)
- Jing-Bo Zhou
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Dongyang Tang
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Lin He
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Shiqi Lin
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Josh Haipeng Lei
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Heng Sun
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Xiaoling Xu
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontier Science Center for Precision Oncology, University of Macau, Macau SAR, China
| | - Chu-Xia Deng
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontier Science Center for Precision Oncology, University of Macau, Macau SAR, China.
| |
Collapse
|
114
|
Tantawy SI, Timofeeva N, Sarkar A, Gandhi V. Targeting MCL-1 protein to treat cancer: opportunities and challenges. Front Oncol 2023; 13:1226289. [PMID: 37601693 PMCID: PMC10436212 DOI: 10.3389/fonc.2023.1226289] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/03/2023] [Indexed: 08/22/2023] Open
Abstract
Evading apoptosis has been linked to tumor development and chemoresistance. One mechanism for this evasion is the overexpression of prosurvival B-cell lymphoma-2 (BCL-2) family proteins, which gives cancer cells a survival advantage. Mcl-1, a member of the BCL-2 family, is among the most frequently amplified genes in cancer. Targeting myeloid cell leukemia-1 (MCL-1) protein is a successful strategy to induce apoptosis and overcome tumor resistance to chemotherapy and targeted therapy. Various strategies to inhibit the antiapoptotic activity of MCL-1 protein, including transcription, translation, and the degradation of MCL-1 protein, have been tested. Neutralizing MCL-1's function by targeting its interactions with other proteins via BCL-2 interacting mediator (BIM)S2A has been shown to be an equally effective approach. Encouraged by the design of venetoclax and its efficacy in chronic lymphocytic leukemia, scientists have developed other BCL-2 homology (BH3) mimetics-particularly MCL-1 inhibitors (MCL-1i)-that are currently in clinical trials for various cancers. While extensive reviews of MCL-1i are available, critical analyses focusing on the challenges of MCL-1i and their optimization are lacking. In this review, we discuss the current knowledge regarding clinically relevant MCL-1i and focus on predictive biomarkers of response, mechanisms of resistance, major issues associated with use of MCL-1i, and the future use of and maximization of the benefits from these agents.
Collapse
Affiliation(s)
- Shady I. Tantawy
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Natalia Timofeeva
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Aloke Sarkar
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Varsha Gandhi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
115
|
Priantti JN, Vilbert M, Madeira T, Moraes FCA, Hein ECK, Saeed A, Cavalcante L. Efficacy and Safety of Rechallenge with BRAF/MEK Inhibitors in Advanced Melanoma Patients: A Systematic Review and Meta-Analysis. Cancers (Basel) 2023; 15:3754. [PMID: 37568570 PMCID: PMC10417341 DOI: 10.3390/cancers15153754] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/20/2023] [Accepted: 07/23/2023] [Indexed: 08/13/2023] Open
Abstract
This systematic review and meta-analysis aims to evaluate the efficacy and safety of rechallenging advanced melanoma patients with BRAFi/MEKi. Seven studies, accounting for 400 patients, were included. Most patients received immunotherapy before the rechallenge, and 79% underwent rechallenge with the combination of BRAFi/MEKi. We found a median progression-free survival of 5 months and overall survival of 9.8 months. The one-year survival rate was 42.63%. Regarding response, ORR was 34% and DCR 65%. There were no new or unexpected safety concerns. Rechallenge with BRAFi/MEKi can improve outcomes in advanced melanoma patients with refractory disease. These findings have significant implications for clinical practice, particularly in the setting of progressive disease in later lines and limited treatment options.
Collapse
Affiliation(s)
- Jonathan N. Priantti
- School of Medicine, Federal University of Amazonas—UFAM, Manaus 69020-160, AM, Brazil
| | - Maysa Vilbert
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
- Division of Medical Oncology, Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Thiago Madeira
- School of Medicine, Federal University of Minas Gerais—UFMG, Belo Horizonte 30130-100, MG, Brazil
| | | | - Erica C. Koch Hein
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
- Division of Medical Oncology, Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Hematology and Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Anwaar Saeed
- Department of Medicine, Division of Hematology and Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Ludimila Cavalcante
- Department of Medical Oncology, Novant Health Cancer Institute, Charlotte, NC 28204, USA
| |
Collapse
|
116
|
Hasanov M, Milton DR, Davies AB, Sirmans E, Saberian C, Posada EL, Opusunju S, Gershenwald JE, Torres-Cabala CA, Burton EM, Colen RR, Huse JT, Glitza Oliva IC, Chung C, McAleer MF, McGovern SL, Yeboa DN, Kim BYS, Prabhu SS, McCutcheon IE, Weinberg JS, Lang FF, Tawbi HA, Li J, Haydu LE, Davies MA, Ferguson SD. Changes in outcomes and factors associated with survival in melanoma patients with brain metastases. Neuro Oncol 2023; 25:1310-1320. [PMID: 36510640 PMCID: PMC10326492 DOI: 10.1093/neuonc/noac251] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Treatment options for patients with melanoma brain metastasis (MBM) have changed significantly in the last decade. Few studies have evaluated changes in outcomes and factors associated with survival in MBM patients over time. The aim of this study is to evaluate changes in clinical features and overall survival (OS) for MBM patients. METHODS Patients diagnosed with MBMs from 1/1/2009 to 12/31/2013 (Prior Era; PE) and 1/1/2014 to 12/31/2018 (Current Era; CE) at The University of Texas MD Anderson Cancer Center were included in this retrospective analysis. The primary outcome measure was OS. Log-rank test assessed differences between groups; multivariable analyses were performed with Cox proportional hazards models and recursive partitioning analysis (RPA). RESULTS A total of 791 MBM patients (PE, n = 332; CE, n = 459) were included in analysis. Median OS from MBM diagnosis was 10.3 months (95% CI, 8.9-12.4) and improved in the CE vs PE (14.4 vs 10.3 months, P < .001). Elevated serum lactate dehydrogenase (LDH) was the only factor associated with worse OS in both PE and CE patients. Factors associated with survival in CE MBM patients included patient age, primary tumor Breslow thickness, prior immunotherapy, leptomeningeal disease, symptomatic MBMs, and whole brain radiation therapy. Several factors associated with OS in the PE were not significant in the CE. RPA demonstrated that elevated serum LDH and prior immunotherapy treatment are the most important determinants of survival in CE MBM patients. CONCLUSIONS OS and factors associated with OS have changed for MBM patients. This information can inform contemporary patient management and clinical investigations.
Collapse
Affiliation(s)
- Merve Hasanov
- Department of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Denái R Milton
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Alicia Bea Davies
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Elizabeth Sirmans
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Chantal Saberian
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Eliza L Posada
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sylvia Opusunju
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Carlos A Torres-Cabala
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Elizabeth M Burton
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rivka R Colen
- Center for Artificial Intelligence Innovation in Medical Imaging, University of Pittsburg, Pittsburg, Pennsylvania, USA
| | - Jason T Huse
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Isabella C Glitza Oliva
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Caroline Chung
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mary Frances McAleer
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Susan L McGovern
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Debra N Yeboa
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sujit S Prabhu
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ian E McCutcheon
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jeffrey S Weinberg
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Frederick F Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hussein A Tawbi
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jing Li
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lauren E Haydu
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael A Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sherise D Ferguson
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
117
|
Marani A, Gioacchini H, Paolinelli M, Offidani A, Campanati A. Potential drug-drug interactions with mitogen-activated protein kinase (MEK) inhibitors used to treat melanoma. Expert Opin Drug Metab Toxicol 2023; 19:555-567. [PMID: 37659065 DOI: 10.1080/17425255.2023.2255519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/08/2023] [Accepted: 09/01/2023] [Indexed: 09/05/2023]
Abstract
INTRODUCTION The management of patients with BRAF-mutated advanced melanoma who are undergoing targeted therapy with MEK inhibitors can be complicated by the co-administration of multiple medications, which can give rise to drug-drug interactions of clinical significance. COVERED AREAS Our review presents a comprehensive analysis of the pharmacokinetic and pharmacodynamic interactions of the three approved for advanced melanoma MEK inhibitor drugs - binimetinib, cobimetinib, and trametinib. MEDLINE (PubMed) was utilized for the literature search, comprising clinical studies, observational studies, and preclinical research. The review discusses the impact of these interactions on efficacy and safety of the treatments and differentiates between interactions supported by pharmacokinetic or pharmacodynamic mechanisms, those encountered in clinical practice, and those observed in preclinical studies. EXPERT OPINION Physicians should be aware about potential benefits, but also increased toxicity caused by drug interactions between MEK inhibitors and other drugs in the management of patients with metastatic melanoma.
Collapse
Affiliation(s)
- A Marani
- Dermatologic Clinic, Department of Clinical and Molecular Sciences, Ancona, Marche, Italy
| | - H Gioacchini
- Dermatologic Clinic, Department of Clinical and Molecular Sciences, Ancona, Marche, Italy
| | - M Paolinelli
- Dermatologic Clinic, Department of Clinical and Molecular Sciences, Ancona, Marche, Italy
| | - A Offidani
- Dermatologic Clinic, Department of Clinical and Molecular Sciences, Ancona, Marche, Italy
| | - A Campanati
- Dermatologic Clinic, Department of Clinical and Molecular Sciences, Ancona, Marche, Italy
| |
Collapse
|
118
|
Das A, Ghose A, Naicker K, Sanchez E, Chargari C, Rassy E, Boussios S. Advances in adoptive T-cell therapy for metastatic melanoma. Curr Res Transl Med 2023; 71:103404. [PMID: 37478776 DOI: 10.1016/j.retram.2023.103404] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/30/2023] [Accepted: 07/06/2023] [Indexed: 07/23/2023]
Abstract
Adoptive T cell therapy (ACT) is a fast developing, niche area of immunotherapy (IO), which is revolutionising the therapeutic landscape of solid tumour oncology, especially metastatic melanoma (MM). Identifying tumour antigens (TAs) as potential targets, the ACT response is mediated by either Tumour Infiltrating Lymphocytes (TILs) or genetically modified T cells with specific receptors - T cell receptors (TCRs) or chimeric antigen receptors (CARs) or more prospectively, natural killer (NK) cells. Clinical trials involving ACT in MM from 2006 to present have shown promising results. Yet it is not without its drawbacks which include significant auto-immune toxicity and need for pre-conditioning lymphodepletion. Although immune-modulation is underway using various combination therapies in the hope of enhancing efficacy and reducing toxicity. Our review article explores the role of ACT in MM, including the various modalities - their safety, efficacy, risks and their development in the trial and the real world setting.
Collapse
Affiliation(s)
- Aparimita Das
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, ME7 5NY, Gillingham, Kent, United Kingdom; Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chennai, India
| | - Aruni Ghose
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, ME7 5NY, Gillingham, Kent, United Kingdom; Department of Medical Oncology, Barts Cancer Centre, St. Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom; Department of Medical Oncology, Mount Vernon Cancer Centre, East and North Hertfordshire NHS Trust, London, United Kingdom
| | - Kevin Naicker
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, ME7 5NY, Gillingham, Kent, United Kingdom
| | - Elisabet Sanchez
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, ME7 5NY, Gillingham, Kent, United Kingdom
| | - Cyrus Chargari
- Department of Radiation Oncology, Pitié Salpêtrière University Hospital, Paris, France
| | - Elie Rassy
- Department of Medical Oncology, Gustave Roussy Institut, 94805, Villejuif, France
| | - Stergios Boussios
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, ME7 5NY, Gillingham, Kent, United Kingdom; Kent and Medway Medical School, University of Kent, Canterbury, United Kingdom; Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King's College London, SE1 9RT, London, United Kingdom; AELIA Organization, 9th Km Thessaloniki, Thermi 57001, Thessaloniki, Greece.
| |
Collapse
|
119
|
Perez MC, Depalo DK, Zager JS. A safety review of recently approved and late-stage trial treatments for metastatic melanoma: systemic and regional therapies. Expert Opin Drug Saf 2023; 22:789-797. [PMID: 37551723 DOI: 10.1080/14740338.2023.2245333] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/30/2023] [Accepted: 08/03/2023] [Indexed: 08/09/2023]
Abstract
INTRODUCTION Advanced melanoma accounts for the majority of skin cancer-associated deaths. Over the past 15 years, there has been a dramatic change in the treatment options and prognosis for patients with advanced melanoma secondary to the development of novel systemic immunotherapies (IO) and targeted therapies. In addition to these novel systemic therapies, regional therapies (intralesional and perfusional) also continue to play a major role in the management of these patients. AREAS COVERED In this article, we review recent updates in the management of advanced melanoma via Medline (PubMed) and Google Scholar, including recently published trials in the metastatic, adjuvant, and neoadjuvant settings. We also review recently published trials for regional therapies and discuss future directions in the management of patients with advanced melanoma. EXPERT OPINION A significant portion of patients with advanced melanoma will develop recurrent or progressive disease following treatment with IO or targeted therapy. Therefore, identifying not only the appropriate therapeutic agent but also the sequence and duration of treatment is pivotal for these patients.
Collapse
Affiliation(s)
- Matthew C Perez
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa FL, United States of America
| | - Danielle K Depalo
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa FL, United States of America
| | - Jonathan S Zager
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa FL, United States of America
| |
Collapse
|
120
|
Kim S, Carvajal R, Kim M, Yang HW. Kinetics of RTK activation determine ERK reactivation and resistance to dual BRAF/MEK inhibition in melanoma. Cell Rep 2023; 42:112570. [PMID: 37252843 DOI: 10.1016/j.celrep.2023.112570] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 03/31/2023] [Accepted: 05/12/2023] [Indexed: 06/01/2023] Open
Abstract
The combination of BRAF and MEK inhibitors (BRAFi/MEKi) has shown promising response rates in treating BRAF-mutant melanoma by inhibiting ERK activation. However, treatment efficacy is limited by the emergence of drug-tolerant persister cells (persisters). Here, we show that the magnitude and duration of receptor tyrosine kinase (RTK) activation determine ERK reactivation and persister development. Our single-cell analysis reveals that only a small subset of melanoma cells exhibits effective RTK and ERK activation and develops persisters, despite uniform external stimuli. The kinetics of RTK activation directly influence ERK signaling dynamics and persister development. These initially rare persisters form major resistant clones through effective RTK-mediated ERK activation. Consequently, limiting RTK signaling suppresses ERK activation and cell proliferation in drug-resistant cells. Our findings provide non-genetic mechanistic insights into the role of heterogeneity in RTK activation kinetics in ERK reactivation and BRAFi/MEKi resistance, suggesting potential strategies for overcoming drug resistance in BRAF-mutant melanoma.
Collapse
Affiliation(s)
- Sungsoo Kim
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Richard Carvajal
- Department of Medicine, Columbia University, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA
| | - Minah Kim
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA
| | - Hee Won Yang
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
121
|
Cucci MA, Grattarola M, Monge C, Roetto A, Barrera G, Caputo E, Dianzani C, Pizzimenti S. Nrf2 as a Therapeutic Target in the Resistance to Targeted Therapies in Melanoma. Antioxidants (Basel) 2023; 12:1313. [PMID: 37372043 DOI: 10.3390/antiox12061313] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/16/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
The use of specific inhibitors towards mutant BRAF (BRAFi) and MEK (MEKi) in BRAF-mutated patients has significantly improved progression-free and overall survival of metastatic melanoma patients. Nevertheless, half of the patients still develop resistance within the first year of therapy. Therefore, understanding the mechanisms of BRAFi/MEKi-acquired resistance has become a priority for researchers. Among others, oxidative stress-related mechanisms have emerged as a major force. The aim of this study was to evaluate the contribution of Nrf2, the master regulator of the cytoprotective and antioxidant response, in the BRAFi/MEKi acquired resistance of melanoma. Moreover, we investigated the mechanisms of its activity regulation and the possible cooperation with the oncogene YAP, which is also involved in chemoresistance. Taking advantage of established in vitro melanoma models resistant to BRAFi, MEKi, or dual resistance to BRAFi/MEKi, we demonstrated that Nrf2 was upregulated in melanoma cells resistant to targeted therapy at the post-translational level and that the deubiquitinase DUB3 participated in the control of the Nrf2 protein stability. Furthermore, we found that Nrf2 controlled the expression of YAP. Importantly, the inhibition of Nrf2, directly or through inhibition of DUB3, reverted the resistance to targeted therapies.
Collapse
Affiliation(s)
- Marie Angèle Cucci
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125 Turin, Italy
| | - Margherita Grattarola
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125 Turin, Italy
| | - Chiara Monge
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy
| | - Antonella Roetto
- Department of Clinical and Biological Sciences-San Luigi Gonzaga Hospital, University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Giuseppina Barrera
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125 Turin, Italy
| | - Emilia Caputo
- Institute of Genetics and Biophysics-IGB-CNR, "A. Buzzati-Traverso", Via Pietro Castellino 111, 80131 Naples, Italy
| | - Chiara Dianzani
- Department of Scienza e Tecnologia del Farmaco, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy
| | - Stefania Pizzimenti
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125 Turin, Italy
| |
Collapse
|
122
|
Subbiah V, Kreitman RJ, Wainberg ZA, Gazzah A, Lassen U, Stein A, Wen PY, Dietrich S, de Jonge MJA, Blay JY, Italiano A, Yonemori K, Cho DC, de Vos FYFL, Moreau P, Fernandez EE, Schellens JHM, Zielinski CC, Redhu S, Boran A, Passos VQ, Ilankumaran P, Bang YJ. Dabrafenib plus trametinib in BRAFV600E-mutated rare cancers: the phase 2 ROAR trial. Nat Med 2023; 29:1103-1112. [PMID: 37059834 PMCID: PMC10202803 DOI: 10.1038/s41591-023-02321-8] [Citation(s) in RCA: 131] [Impact Index Per Article: 65.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/27/2023] [Indexed: 04/16/2023]
Abstract
BRAFV600E alterations are prevalent across multiple tumors. Here we present final efficacy and safety results of a phase 2 basket trial of dabrafenib (BRAF kinase inhibitor) plus trametinib (MEK inhibitor) in eight cohorts of patients with BRAFV600E-mutated advanced rare cancers: anaplastic thyroid carcinoma (n = 36), biliary tract cancer (n = 43), gastrointestinal stromal tumor (n = 1), adenocarcinoma of the small intestine (n = 3), low-grade glioma (n = 13), high-grade glioma (n = 45), hairy cell leukemia (n = 55) and multiple myeloma (n = 19). The primary endpoint of investigator-assessed overall response rate in these cohorts was 56%, 53%, 0%, 67%, 54%, 33%, 89% and 50%, respectively. Secondary endpoints were median duration of response (DoR), progression-free survival (PFS), overall survival (OS) and safety. Median DoR was 14.4 months, 8.9 months, not reached, 7.7 months, not reached, 31.2 months, not reached and 11.1 months, respectively. Median PFS was 6.7 months, 9.0 months, not reached, not evaluable, 9.5 months, 5.5 months, not evaluable and 6.3 months, respectively. Median OS was 14.5 months, 13.5 months, not reached, 21.8 months, not evaluable, 17.6 months, not evaluable and 33.9 months, respectively. The most frequent (≥20% of patients) treatment-related adverse events were pyrexia (40.8%), fatigue (25.7%), chills (25.7%), nausea (23.8%) and rash (20.4%). The encouraging tumor-agnostic activity of dabrafenib plus trametinib suggests that this could be a promising treatment approach for some patients with BRAFV600E-mutated advanced rare cancers. ClinicalTrials.gov registration: NCT02034110 .
Collapse
Affiliation(s)
- Vivek Subbiah
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Robert J Kreitman
- Laboratory of Molecular Biology, National Institutes of Health, Bethesda, MD, USA
| | - Zev A Wainberg
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Anas Gazzah
- Drug Development Department (DITEP), Gustave Roussy Cancer Institute, Villejuif, France
| | - Ulrik Lassen
- Department of Oncology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Alexander Stein
- Department of Internal Medicine II (Oncology Center), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Maja J A de Jonge
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Jean-Yves Blay
- Center Leon Berard & University Claude Bernard Lyon I, Lyon, France
| | - Antoine Italiano
- Early Phase Trials and Sarcoma Units, Institut Bergonié, Bordeaux, France; Faculty of Medicine, University of Bordeaux, Bordeaux, France
| | | | | | - Filip Y F L de Vos
- Department of Medical Oncology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | | | - Elena Elez Fernandez
- Department of Medical Oncology, Vall d'Hebron University Hospital (HUVH), Vall d'Hebron Institute of Oncology (VHIO), IOB-Quiron, UVic-UCC, Barcelona, Spain
| | | | | | - Suman Redhu
- Global Program Biostatistics, Novartis Oncology, Cambridge, MA, USA
| | - Aislyn Boran
- Global Drug Development, Oncology Development Unit, Novartis Services, Inc., East Hanover, NJ, USA
| | - Vanessa Q Passos
- Global Drug Development, Oncology Development Unit, Novartis Services, Inc., East Hanover, NJ, USA
| | - Palanichamy Ilankumaran
- Global Drug Development, Oncology Development Unit, Novartis Services, Inc., East Hanover, NJ, USA
| | - Yung-Jue Bang
- Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
123
|
Limberg J, Egan CE, Gray KD, Singh M, Loewenstein Z, Yang Y, Riascos MC, Al Asadi H, Safe P, El Eshaky S, Liang H, Ullmann TM, Wang W, Li W, Zhang T, Xiang J, Stefanova D, Jin MM, Zarnegar R, Fahey TJ, Min IM. Activation of the JAK/STAT Pathway Leads to BRAF Inhibitor Resistance in BRAFV600E Positive Thyroid Carcinoma. Mol Cancer Res 2023; 21:397-410. [PMID: 36790391 PMCID: PMC10159921 DOI: 10.1158/1541-7786.mcr-21-0832] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 03/25/2022] [Accepted: 01/11/2023] [Indexed: 02/16/2023]
Abstract
A subset of thyroid cancers, recurrent differentiated thyroid cancers and anaplastic thyroid cancer (ATC), are difficult to treat by thyroidectomy and systemic therapy. A common mutation in thyroid cancer, BRAFV600E, has targetable treatment options; however, the results have been disappointing in thyroid cancers compared with BRAFV600E melanoma, as thyroid cancers quickly become resistant to BRAFV600E inhibitor (BRAFi). Here, we studied the molecular pathway that is induced in BRAFV600E thyroid cancer cells and patient-derived tumor samples in response to BRAFi, vemurafenib, using RNA-sequencing and molecular analysis. Both inducible response to BRAFi and acquired BRAFi resistance in BRAFV600E thyroid cancer cells showed significant activation of the JAK/STAT pathway. Functional analyses revealed that the combination of BRAFi and inhibitors of JAK/STAT pathway controlled BRAFV600E thyroid cancer cell growth. The Cancer Genome Atlas data analysis demonstrated that potent activation of the JAK/STAT signaling was associated with shorter recurrence rate in patients with differentiated thyroid cancer. Analysis of tumor RNA expression in patients with poorly differentiated thyroid cancer and ATC also support that enhanced activity of JAK/STAT signaling pathway is correlated with worse prognosis. Our study demonstrates that JAK/STAT pathway is activated as BRAFV600E thyroid cancer cells develop resistance to BRAFi and that this pathway is a potential target for anticancer activity and to overcome drug resistance that commonly develops to treatment with BRAFi in thyroid cancer. IMPLICATIONS Dual inhibition of BRAF and JAK/STAT signaling pathway is a potential therapeutic treatment for anticancer activity and to overcome drug resistance to BRAFi in thyroid cancer.
Collapse
Affiliation(s)
- Jessica Limberg
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | - Caitlin E. Egan
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | | | - Mandeep Singh
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | | | - Yanping Yang
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | | | - Hala Al Asadi
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | - Parima Safe
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | - Steve El Eshaky
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | - Heng Liang
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | | | - Weibin Wang
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | - Wei Li
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | - Tuo Zhang
- Genomics Resource Core Facility, Weill Cornell Medicine, New York, NY 10065
| | - Jenny Xiang
- Genomics Resource Core Facility, Weill Cornell Medicine, New York, NY 10065
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10065
| | | | - Moonsoo M. Jin
- Department of Radiology, Weill Cornell Medicine, New York, NY 10065
| | - Rasa Zarnegar
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | - Thomas J. Fahey
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| | - Irene M. Min
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065
| |
Collapse
|
124
|
Neuendorf HM, Simmons JL, Boyle GM. Therapeutic targeting of anoikis resistance in cutaneous melanoma metastasis. Front Cell Dev Biol 2023; 11:1183328. [PMID: 37181747 PMCID: PMC10169659 DOI: 10.3389/fcell.2023.1183328] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 04/14/2023] [Indexed: 05/16/2023] Open
Abstract
The acquisition of resistance to anoikis, the cell death induced by loss of adhesion to the extracellular matrix, is an absolute requirement for the survival of disseminating and circulating tumour cells (CTCs), and for the seeding of metastatic lesions. In melanoma, a range of intracellular signalling cascades have been identified as potential drivers of anoikis resistance, however a full understanding of the process is yet to be attained. Mechanisms of anoikis resistance pose an attractive target for the therapeutic treatment of disseminating and circulating melanoma cells. This review explores the range of small molecule, peptide and antibody inhibitors targeting molecules involved in anoikis resistance in melanoma, and may be repurposed to prevent metastatic melanoma prior to its initiation, potentially improving the prognosis for patients.
Collapse
Affiliation(s)
- Hannah M. Neuendorf
- Cancer Drug Mechanisms Group, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Jacinta L. Simmons
- Cancer Drug Mechanisms Group, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Glen M. Boyle
- Cancer Drug Mechanisms Group, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
125
|
Tateo V, Marchese PV, Mollica V, Massari F, Kurzrock R, Adashek JJ. Agnostic Approvals in Oncology: Getting the Right Drug to the Right Patient with the Right Genomics. Pharmaceuticals (Basel) 2023; 16:ph16040614. [PMID: 37111371 PMCID: PMC10144220 DOI: 10.3390/ph16040614] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
(1) Background: The oncology field has drastically changed with the advent of precision medicine, led by the discovery of druggable genes or immune targets assessed through next-generation sequencing. Biomarker-based treatments are increasingly emerging, and currently, six tissue-agnostic therapies are FDA-approved. (2) Methods: We performed a review of the literature and reported the trials that led to the approval of tissue-agnostic treatments and ongoing clinical trials currently investigating novel biomarker-based approaches. (3) Results: We discussed the approval of agnostic treatments: pembrolizumab and dostarlimab for MMRd/MSI-H, pembrolizumab for TMB-H, larotrectinib and entrectinib for NTRK-fusions, dabrafenib plus trametinib for BRAF V600E mutation, and selpercatinib for RET fusions. In addition, we reported novel clinical trials of biomarker-based approaches, including ALK, HER2, FGFR, and NRG1. (4) Conclusions: Precision medicine is constantly evolving, and with the improvement of diagnostic tools that allow a wider genomic definition of the tumor, tissue-agnostic targeted therapies are a promising treatment strategy tailored to the specific tumor genomic profile, leading to improved survival outcomes.
Collapse
Affiliation(s)
- Valentina Tateo
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Paola Valeria Marchese
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40127 Bologna, Italy
| | - Razelle Kurzrock
- MCW Cancer Center, Milwaukee, WI 53226, USA
- WIN Consortium, San Diego, CA 92093, USA
- Department of Oncology, University of Nebraska, Omaha, NE 68198, USA
| | - Jacob J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD 21287, USA
| |
Collapse
|
126
|
Said SS, Ibrahim WN. Cancer Resistance to Immunotherapy: Comprehensive Insights with Future Perspectives. Pharmaceutics 2023; 15:pharmaceutics15041143. [PMID: 37111629 PMCID: PMC10141036 DOI: 10.3390/pharmaceutics15041143] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/24/2023] [Accepted: 03/31/2023] [Indexed: 04/08/2023] Open
Abstract
Cancer immunotherapy is a type of treatment that harnesses the power of the immune systems of patients to target cancer cells with better precision compared to traditional chemotherapy. Several lines of treatment have been approved by the US Food and Drug Administration (FDA) and have led to remarkable success in the treatment of solid tumors, such as melanoma and small-cell lung cancer. These immunotherapies include checkpoint inhibitors, cytokines, and vaccines, while the chimeric antigen receptor (CAR) T-cell treatment has shown better responses in hematological malignancies. Despite these breakthrough achievements, the response to treatment has been variable among patients, and only a small percentage of cancer patients gained from this treatment, depending on the histological type of tumor and other host factors. Cancer cells develop mechanisms to avoid interacting with immune cells in these circumstances, which has an adverse effect on how effectively they react to therapy. These mechanisms arise either due to intrinsic factors within cancer cells or due other cells within the tumor microenvironment (TME). When this scenario is used in a therapeutic setting, the term “resistance to immunotherapy” is applied; “primary resistance” denotes a failure to respond to treatment from the start, and “secondary resistance” denotes a relapse following the initial response to immunotherapy. Here, we provide a thorough summary of the internal and external mechanisms underlying tumor resistance to immunotherapy. Furthermore, a variety of immunotherapies are briefly discussed, along with recent developments that have been employed to prevent relapses following treatment, with a focus on upcoming initiatives to improve the efficacy of immunotherapy for cancer patients.
Collapse
Affiliation(s)
- Sawsan Sudqi Said
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Wisam Nabeel Ibrahim
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
127
|
Rezaie Y, Fattahi F, Mashinchi B, Kamyab Hesari K, Montazeri S, Kalantari E, Madjd Z, Saeednejad Zanjani L. High expression of Talin-1 is associated with tumor progression and recurrence in melanoma skin cancer patients. BMC Cancer 2023; 23:302. [PMID: 37013489 PMCID: PMC10069040 DOI: 10.1186/s12885-023-10771-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 03/26/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND Talin-1 as a component of multi-protein adhesion complexes plays a role in tumor formation and migration in various malignancies. This study investigated Talin-1 in protein levels as a potential prognosis biomarker in skin tumors. METHODS Talin-1 was evaluated in 106 skin cancer (33 melanomas and 73 non-melanomas skin cancer (NMSC)) and 11 normal skin formalin-fixed paraffin-embedded (FFPE) tissue samples using immunohistochemical technique on tissue microarrays (TMAs). The association between the expression of Talin-1 and clinicopathological parameters, as well as survival outcomes, were assessed. RESULTS Our findings from data minings through bioinformatics tools indicated dysregulation of Talin-1 in mRNA levels for skin cancer samples. In addition, there was a statistically significant difference in Talin-1 expression in terms of intensity of staining, percentage of positive tumor cells, and H-score in melanoma tissues compared to NMSC (P = 0.001, P < 0.001, and P < 0.001, respectively). Moreover, high cytoplasmic expression of Talin-1 was found to be associated with significantly advanced stages (P = 0.024), lymphovascular invasion (P = 0.023), and recurrence (P = 0.006) in melanoma cancer tissues. Our results on NMSC showed a statistically significant association between high intensity of staining and the poor differentiation (P = 0.044). No significant associations were observed between Talin-1 expression levels and survival outcomes of melanoma and NMSC patients. CONCLUSION Our observations showed that higher expression of Talin1 in protein level may be significantly associated with more aggressive tumor behavior and advanced disease in patients with skin cancer. However, further studies are required to find the mechanism of action of Talin-1 in skin cancers.
Collapse
Affiliation(s)
- Yasaman Rezaie
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14535, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Fattahi
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14535, Iran
| | - Baharnaz Mashinchi
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14535, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kambiz Kamyab Hesari
- Department of Dermatopathology, Razi Hospital, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Sahar Montazeri
- Department of Dermatopathology, Razi Hospital, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Elham Kalantari
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14535, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14535, Iran.
| | - Leili Saeednejad Zanjani
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next to Milad Tower, Tehran, 14496-14535, Iran.
- Department of Pathology and Genomic Medicine, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
128
|
Hendrikse CSE, Theelen PMM, van der Ploeg P, Westgeest HM, Boere IA, Thijs AMJ, Ottevanger PB, van de Stolpe A, Lambrechts S, Bekkers RLM, Piek JMJ. The potential of RAS/RAF/MEK/ERK (MAPK) signaling pathway inhibitors in ovarian cancer: A systematic review and meta-analysis. Gynecol Oncol 2023; 171:83-94. [PMID: 36841040 DOI: 10.1016/j.ygyno.2023.01.038] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/01/2023] [Accepted: 01/30/2023] [Indexed: 02/27/2023]
Abstract
BACKGROUND The RAS/RAF/MEK/ERK (MAPK) pathway plays a role in ovarian carcinogenesis. Low-grade serous ovarian carcinoma (LGSOC) frequently harbors activating MAPK mutations. MAPK inhibitors have been used in small subsets of ovarian carcinoma (OC) patients to control tumor growth. Therefore, we performed a meta-analysis to evaluate the effectiveness of MAPK inhibitors in OC patients. We aimed to determine the clinical benefit rate (CBR), the subgroup of MAPK inhibitors with the best CBR and overall response rate (ORR), and the most common adverse events. METHODS We conducted a search in PubMed, Embase via Ovid, the Cochrane library and clinicaltrials.gov on studies evaluating the efficacy of single MAPK pathway inhibition with MAPK pathway inhibitors in OC patients. Our primary outcome included the CBR, defined by the proportion of patients with stable disease (SD), complete (CR) and partial response (PR). Secondary outcomes included the ORR (including PR and CR) and grade 3 and 4 adverse events. Meta-analysis was performed using a random-effects model. RESULTS We included nine studies with a total of 319 OC patients, for which we determined a pooled CBR of 63% (95%-CI 39-84%, I2 = 92%). Combined treatment with Raf- and MEK inhibitors in in BRAFv600 mutated LGSOC (n = 6) had the greatest efficacy with a CBR of 100% and ORR of 83%. MEK inhibitors had the best efficacy as a single agent. Subgroup analysis by tumor histology demonstrated a significantly higher CBR and ORR in patients with LGSOC, with a pooled CBR and ORR of 87% (95%-CI 81-92%, I2 = 0%) and 27% (95%-CI 10-48%, I2 = 77%) respectively. Adverse events of grade 3 or higher were reported frequently: 123 in 167 patients. CONCLUSIONS MEK inhibitors are the most promising single agents in (LGS)OC. However, dual MAPK pathway inhibition should be considered in patients with a BRAFv600 mutation, or non-mutated OC with depleted treatment options due indications of higher efficacy and tolerable toxicity profiles.
Collapse
Affiliation(s)
- C S E Hendrikse
- Department of Obstetrics and Gynecology and Catharina Cancer Institute, Catharina Hospital, Eindhoven, the Netherlands; GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands.
| | - P M M Theelen
- Department of Obstetrics and Gynecology and Catharina Cancer Institute, Catharina Hospital, Eindhoven, the Netherlands
| | - P van der Ploeg
- Department of Obstetrics and Gynecology and Catharina Cancer Institute, Catharina Hospital, Eindhoven, the Netherlands; GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - H M Westgeest
- Department of Internal Medicine, Amphia Hospital, Breda, the Netherlands
| | - I A Boere
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - A M J Thijs
- Department of Internal Medicine and Catharina Cancer Institute, Catharina Hospital, Eindhoven, the Netherlands
| | - P B Ottevanger
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - A van de Stolpe
- Drug Companion Diagnostics Company - Therapeutics (DCDC-Tx), Vught, the Netherlands
| | - S Lambrechts
- Department of Obstetrics and Gynecology, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - R L M Bekkers
- Department of Obstetrics and Gynecology and Catharina Cancer Institute, Catharina Hospital, Eindhoven, the Netherlands; GROW School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - J M J Piek
- Department of Obstetrics and Gynecology and Catharina Cancer Institute, Catharina Hospital, Eindhoven, the Netherlands
| |
Collapse
|
129
|
Henderson CJ, McLaren AW, Kapelyukh Y, Wolf CR. Improving the predictive power of xenograft and syngeneic anti-tumour studies using mice humanised for pathways of drug metabolism. F1000Res 2023; 11:1081. [PMID: 37065929 PMCID: PMC10090862 DOI: 10.12688/f1000research.122987.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/07/2023] [Indexed: 03/29/2023] Open
Abstract
Drug development is an expensive and time-consuming process, with only a small fraction of drugs gaining regulatory approval from the often many thousands of candidates identified during target validation. Once a lead compound has been identified and optimised, they are subject to intensive pre-clinical research to determine their pharmacodynamic, pharmacokinetic and toxicological properties, procedures which inevitably involve significant numbers of animals - mainly mice and rats, but also dogs and monkeys in much smaller numbers and for specific types of drug candidates. Many compounds that emerge from this process, having been shown to be safe and efficacious in pre-clinical studies, subsequently fail to replicate this outcome in clinical trials, therefore wasting time, money and, most importantly, animals. Due to high rates of metabolism and a differing spectrum of metabolites (some pharmacologically active) in rodents, species differences in drug metabolism can be a major impediment to drug discovery programmes and confound the extrapolation of animal data to humans. To circumvent this, we have developed a complex transgenic mouse model – 8HUM - which faithfully replicates human Phase I drug metabolism (and its regulation), and which will generate more human-relevant data from fewer animals in a pre-clinical setting and reduce attrition in the clinic. One key area for the pre-clinical application of animals in an oncology setting – almost exclusively mice - is their use in anti-tumour studies. We now further demonstrate the utility of the 8HUM mouse using a murine melanoma cell line as a syngeneic tumour and also present an immunodeficient version 8HUM_Rag2 -/- - for use in xenograft studies. These models will be of significant benefit not only to Pharma for pre-clinical drug development work, but also throughout the drug efficacy, toxicology, pharmacology, and drug metabolism communities, where fewer animals will be needed to generate more human-relevant data.
Collapse
Affiliation(s)
- Colin J. Henderson
- Division of Systems Medicine, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, Tayside, DD1 9SY, UK
| | - Aileen W. McLaren
- Division of Systems Medicine, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, Tayside, DD1 9SY, UK
| | - Yury Kapelyukh
- Division of Systems Medicine, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, Tayside, DD1 9SY, UK
| | - C. Roland Wolf
- Division of Systems Medicine, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, Tayside, DD1 9SY, UK
| |
Collapse
|
130
|
Jannelli G, Calvanese F, Paun L, Raverot G, Jouanneau E. Current Advances in Papillary Craniopharyngioma: State-Of-The-Art Therapies and Overview of the Literature. Brain Sci 2023; 13:515. [PMID: 36979325 PMCID: PMC10046497 DOI: 10.3390/brainsci13030515] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/13/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023] Open
Abstract
Craniopharyngiomas are commonly classified as low-grade tumors, although they may harbor a malignant behavior due to their high rate of recurrence and long-term morbidity. Craniopharyngiomas are classically distinguished into two histological types (adamantinomatous and papillary), which have been recently considered by the WHO classification of CNS tumors as two independent entities, due to different epidemiological, radiological, histopathological, and genetic patterns. With regard to papillary craniopharyngioma, a BRAF V600 mutation is detected in 95% of cases. This genetic feature is opening new frontiers in the treatment of these tumors using an adjuvant or, in selected cases, a neo-adjuvant approach. In this article, we present an overview of the more recent literature, focusing on the specificities and the role of oncological treatment in the management of papillary craniopharyngiomas. Based on our research and experience, we strongly suggest a multimodal approach combining clinical, endocrinological, radiological, histological, and oncological findings in both preoperative workup and postoperative follow up to define a roadmap integrating every aspect of this challenging condition.
Collapse
Affiliation(s)
- Gianpaolo Jannelli
- Skull Base and Pituitary Unit, Department of Neurosurgery B, Neurological Hospital Pierre Wertheimer, Bron, 69677 Lyon, France; (G.J.)
- Neurosurgical Unit, Faculty of Medicine, Geneva University Hospitals, University of Geneva, 1205 Geneva, Switzerland
| | - Francesco Calvanese
- Skull Base and Pituitary Unit, Department of Neurosurgery B, Neurological Hospital Pierre Wertheimer, Bron, 69677 Lyon, France; (G.J.)
- Department of Neurosurgery, Helsinki University Central Hospital, Helsinki University, Meilahden tornisairaala, Haartmaninkatu 4 Rakennus 1, 00290 Helsinki, Finland
| | - Luca Paun
- Neurosurgical Unit, Faculty of Medicine, Geneva University Hospitals, University of Geneva, 1205 Geneva, Switzerland
- Department of Neurosurgery, GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, 1 Rue Cabanis, CEDEX 14, 75014 Paris, France
| | - Gerald Raverot
- Department of Endocrinology, Neurological Hospital Pierre Wertheimer, University Hospital of Lyon, 69500 Lyon, France
- Inserm U1052, CNRS UMR5286, Cancer Research Center of Lyon, University Claude Bernard Lyon 1, 69000 Lyon, France
| | - Emmanuel Jouanneau
- Skull Base and Pituitary Unit, Department of Neurosurgery B, Neurological Hospital Pierre Wertheimer, Bron, 69677 Lyon, France; (G.J.)
- Inserm U1052, CNRS UMR5286, Cancer Research Center of Lyon, University Claude Bernard Lyon 1, 69000 Lyon, France
| |
Collapse
|
131
|
Diaz MJ, Mark I, Rodriguez D, Gelman B, Tran JT, Kleinberg G, Levin A, Beneke A, Root KT, Tran AXV, Lucke-Wold B. Melanoma Brain Metastases: A Systematic Review of Opportunities for Earlier Detection, Diagnosis, and Treatment. Life (Basel) 2023; 13:828. [PMID: 36983983 PMCID: PMC10053844 DOI: 10.3390/life13030828] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/25/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Introduction: Melanoma continues to represent the most serious skin cancer worldwide. However, few attempts have been made to connect the body of research on advanced melanoma. In the present review, we report on strides made in the diagnosis and treatment of intracranial metastatic melanoma. Methods: Relevant Cochrane reviews and randomized-controlled trials published by November 2022 were systematically retrieved from the Cochrane Library, EMBASE, and PubMed databases (N = 27). Search and screening methods adhered to the 2020 revision of the Preferred Reporting Items for Systematic reviews and Meta-Analyses guidelines. Results: Although the research surrounding the earlier detection of melanoma brain metastasis is scarce, several studies have highlighted specific markers associated with MBM. Such factors include elevated BRAFV600 mutant ctDNA, high LDH concentration, and high IGF-1R. The approach to treating MBM is moving away from surgery and toward nonsurgical management, namely, a combination of stereotactic radiosurgery (SRS) and immunotherapeutic agents. There is an abundance of emerging research seeking to identify and improve both novel and established treatment options and diagnostic approaches for MBM, however, more research is still needed to maximize the clinical efficacy, especially for new immunotherapeutics. Conclusions: Early detection is optimal for the efficacy of treatment and MBM prognosis. Current treatment utilizes chemotherapies and targeted therapies. Emerging approaches emphasize biomarkers and joint treatments. Further exploration toward preliminary identification, the timing of therapies, and methods to ameliorate adverse treatment effects are needed to advance MBM patient care.
Collapse
Affiliation(s)
| | - Isabella Mark
- College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Daphnee Rodriguez
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Beata Gelman
- Department of Biology, University of Maryland, College Park, MD 20742, USA
| | - Jasmine Thuy Tran
- School of Medicine, University of Indiana, Indianapolis, IN 46202, USA
| | - Giona Kleinberg
- College of Engineering, Northeastern University, Boston, MA 02115, USA
| | - Anna Levin
- School of Arts and Sciences, Rutgers University, Piscataway, NJ 08854, USA
| | - Alice Beneke
- College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Kevin Thomas Root
- College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Andrew Xuan Vinh Tran
- Department of Dermatology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
132
|
Piejko K, Cybulska-Stopa B, Ziętek M, Dziura R, Galus Ł, Kempa-Kamińska N, Ziółkowska B, Rutkowska E, Kopciński T, Kubiatowski T, Bal W, Suwiński R, Mackiewicz J, Kamińska-Winciorek G, Czarnecka AM, Rutkowski P. Long-Term Real-World Outcomes and Safety of Vemurafenib and Vemurafenib + Cobimetinib Therapy in Patients with BRAF-Mutated Melanoma. Target Oncol 2023; 18:235-245. [PMID: 36906728 PMCID: PMC10042754 DOI: 10.1007/s11523-023-00954-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2023] [Indexed: 03/13/2023]
Abstract
BACKGROUND Combined treatment with BRAFi and/or MEK inhibitors (MEKi) improves outcomes in advanced melanoma patients in comparison with monotherapy. OBJECTIVE We aim to report real-world treatment efficacy and safety of vemurafenib (V) and vemurafenib + cobimetinib (V + C) from 10 years of practice. PATIENTS AND METHODS A total of 275 consecutive patients with unresectable or metastatic BRAF mutated melanoma started first-line V or V + C treatment between 1 October 2013 and 31 December 2020. Survival analyses were performed using the Kaplan-Meier method, and Log-rank and Chi-square tests were used for comparison between groups. RESULTS The estimated median overall survival (mOS) was 10.3 months in the V group, and 12.3 months in the V + C group (p = 0.0005; HR = 1.58, 95% CI 1.2-2.1), although the latter group of patients had lactate dehydrogenase elevated numerically more often. Estimated median progression-free survival (mPFS) was 5.5 months in the V group, and 8.3 months in the V + C group (p = 0.0002; HR = 1.62, 95% CI 1.3-2.1). Complete response, partial response, stable disease, and progressive disease as best responses were recorded in the V/V + C groups in 7%/10%, 52%/46%, 26%/28%, and 15%/16% of patients, respectively. The numbers of patients with any grade of adverse effects were similar in both groups. CONCLUSIONS We confirmed significant improvement in the mOS and mPFS of unresectable and/or metastatic BRAF mutated-melanoma patients treated outside clinical trials with V + C as compared with V, with no major increase in toxicity for the combination.
Collapse
Affiliation(s)
- Karolina Piejko
- Department of Clinical Oncology, Clinical Oncology Clinic, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow Branch, Garncarska 11, 31-115, Cracow, Poland
| | - Bożena Cybulska-Stopa
- Department of Clinical Oncology, Clinical Oncology Clinic, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow Branch, Garncarska 11, 31-115, Cracow, Poland. .,Department of Clinical Oncology, Wroclaw Comprehensive Cancer Center, Wroclaw, Poland.
| | - Marcin Ziętek
- Department of Surgical Oncology, Wroclaw Comprehensive Cancer Center, Wroclaw, Poland.,Department of Oncology, Wroclaw Medical University, Wroclaw, Poland
| | - Robert Dziura
- Clinical Oncology Department, Holy Cross Cancer Center, Kielce, Poland
| | - Łukasz Galus
- Department of Medical and Experimental Oncology, University of Medical Sciences, Poznan, Poland
| | | | - Barbara Ziółkowska
- II Clinic of Radiotherapy and Chemotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Ewa Rutkowska
- Clinical Oncology Department, Holy Cross Cancer Center, Kielce, Poland
| | - Tomasz Kopciński
- Department of Clinical Oncology, Clinical Oncology Clinic, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow Branch, Garncarska 11, 31-115, Cracow, Poland
| | - Tomasz Kubiatowski
- Warmian-Masurian Cancer Center of The Ministry of The Interior and Administration's Hospital, Olsztyn, Poland
| | - Wiesław Bal
- Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Rafał Suwiński
- II Clinic of Radiotherapy and Chemotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Jacek Mackiewicz
- Department of Medical and Experimental Oncology, University of Medical Sciences, Poznan, Poland.,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, Poznan, Poland
| | - Grażyna Kamińska-Winciorek
- Department of Bone Marrow Transplantation and Hematology-Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
| | - Anna M Czarnecka
- Clinical Oncology Department, Holy Cross Cancer Center, Kielce, Poland.,Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland.,Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| |
Collapse
|
133
|
Casadonte R, Kriegsmann M, Kriegsmann K, Streit H, Meliß RR, Müller CSL, Kriegsmann J. Imaging Mass Spectrometry for the Classification of Melanoma Based on BRAF/ NRAS Mutational Status. Int J Mol Sci 2023; 24:ijms24065110. [PMID: 36982192 PMCID: PMC10049262 DOI: 10.3390/ijms24065110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/22/2023] [Accepted: 03/02/2023] [Indexed: 03/30/2023] Open
Abstract
Mutations of the oncogenes v-raf murine sarcoma viral oncogene homolog B1 (BRAF) and neuroblastoma RAS viral oncogene homolog (NRAS) are the most frequent genetic alterations in melanoma and are mutually exclusive. BRAF V600 mutations are predictive for response to the two BRAF inhibitors vemurafenib and dabrafenib and the mitogen-activated protein kinase kinase (MEK) inhibitor trametinib. However, inter- and intra-tumoral heterogeneity and the development of acquired resistance to BRAF inhibitors have important clinical implications. Here, we investigated and compared the molecular profile of BRAF and NRAS mutated and wildtype melanoma patients' tissue samples using imaging mass spectrometry-based proteomic technology, to identify specific molecular signatures associated with the respective tumors. SCiLSLab and R-statistical software were used to classify peptide profiles using linear discriminant analysis and support vector machine models optimized with two internal cross-validation methods (leave-one-out, k-fold). Classification models showed molecular differences between BRAF and NRAS mutated melanoma, and identification of both was possible with an accuracy of 87-89% and 76-79%, depending on the respective classification method applied. In addition, differential expression of some predictive proteins, such as histones or glyceraldehyde-3-phosphate-dehydrogenase, correlated with BRAF or NRAS mutation status. Overall, these findings provide a new molecular method to classify melanoma patients carrying BRAF and NRAS mutations and help provide a broader view of the molecular characteristics of these patients that may help understand the signaling pathways and interactions involving the altered genes.
Collapse
Affiliation(s)
| | - Mark Kriegsmann
- Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Institute of Pathology Wiesbaden, 69120 Heidelberg, Germany
| | - Katharina Kriegsmann
- Department of Hematology Oncology and Rheumatology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Helene Streit
- Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University, 3500 Krems, Austria
| | | | - Cornelia S L Müller
- MVZ für Histologie, Zytologie und Molekulare Diagnostik Trier, 54296 Trier, Germany
| | - Joerg Kriegsmann
- Proteopath GmbH, 54296 Trier, Germany
- Department of Medicine, Faculty of Medicine and Dentistry, Danube Private University, 3500 Krems, Austria
- MVZ für Histologie, Zytologie und Molekulare Diagnostik Trier, 54296 Trier, Germany
| |
Collapse
|
134
|
Egeler MD, van Leeuwen M, Fraterman I, van den Heuvel NMJ, Boekhout AH, Lai-Kwon J, Wilthagen EA, Eriksson H, Haanen JB, Wilgenhof S, Ascierto PA, van Akkooi ACJ, van de Poll-Franse LV. Common toxicities associated with immune checkpoint inhibitors and targeted therapy in the treatment of melanoma: A systematic scoping review. Crit Rev Oncol Hematol 2023; 183:103919. [PMID: 36736511 DOI: 10.1016/j.critrevonc.2023.103919] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/23/2022] [Accepted: 01/20/2023] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION This systematic scoping review compares the toxicities experienced by patients receiving immune checkpoint inhibitors (ICIs) or targeted therapy (TT) for stage III (resected and unresectable) and stage IV melanoma. METHODS OVID Medline, Embase, and PsycInfo were searched to identify Phase III trials reporting toxicities of FDA-approved ICIs and TT for advanced melanoma. AEs that were reported by ≥ 10% of patients in the evaluated trials were included. RESULTS Toxicity profiles of 11208 patients from 24 studies were reviewed. The rate of AEs was lower with ICIs compared to TT. However, ICIs were associated with higher rates of long-term or permanent AEs compared to TT, where toxicities generally were shortterm and reversible with treatment discontinuation. CONCLUSION The toxicity profiles of ICIs and TT vary substantially. Whilst the rate of AEs was lower with ICIs than during TT, it was also associated with higher rates of potentially chronic AEs.
Collapse
Affiliation(s)
- Mees D Egeler
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - Marieke van Leeuwen
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Itske Fraterman
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Noelle M J van den Heuvel
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Annelies H Boekhout
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Julia Lai-Kwon
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Erica A Wilthagen
- Scientific Information Service, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Hanna Eriksson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Medical Unit Head-Neck-, Lung-, Skin Cancer, Skin Cancer Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - John B Haanen
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sofie Wilgenhof
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Paolo A Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Napoli, Italy
| | - Alexander C J van Akkooi
- Melanoma Institute Australia, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Lonneke V van de Poll-Franse
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Research & Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, the Netherlands; Department of Medical and Clinical Psychology, Center of Research on Psychology in Somatic diseases (CoRPS), Tilburg University, Tilburg, the Netherlands
| |
Collapse
|
135
|
Fudaba H, Wakimoto H. Oncolytic virus therapy for malignant gliomas: entering the new era. Expert Opin Biol Ther 2023; 23:269-282. [PMID: 36809883 DOI: 10.1080/14712598.2023.2184256] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
INTRODUCTION To overcome the challenge of treating malignant brain tumors, oncolytic viruses (OVs) represent an innovative therapeutic approach, featuring unique mechanisms of action. The recent conditional approval of the oncolytic herpes simplex virus G47Δ as a therapeutic for malignant brain tumors marked a significant milestone in the long history of OV development in neuro-oncology. AREAS COVERED This review summarizes the results of recently completed and active clinical studies that investigate the safety and efficacy of different OV types in patients with malignant gliomas. The changing landscape of the OV trial design includes expansion of subjects to newly diagnosed tumors and pediatric populations. A variety of delivery methods and new routes of administration are vigorously tested to optimize tumor infection and overall efficacy. New therapeutic strategies such as combination with immunotherapies are proposed that take advantage of the characteristics of OV therapy as an immunotherapy. Preclinical studies of OV have been active and aim to translate new OV strategies to the clinic. EXPERT OPINION For the next decade, clinical trials and preclinical and translational research will continue to drive the development of innovative OV treatments for malignant gliomas and benefit patients and define new OV biomarkers.
Collapse
Affiliation(s)
- Hirotaka Fudaba
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Department of Neurosurgery, Oita University Faculty of Medicine, Yufu, Japan
| | - Hiroaki Wakimoto
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
136
|
Current Trends in Mucosal Melanomas: An Overview. Cancers (Basel) 2023; 15:cancers15051356. [PMID: 36900152 PMCID: PMC10000120 DOI: 10.3390/cancers15051356] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/14/2023] [Accepted: 02/18/2023] [Indexed: 02/25/2023] Open
Abstract
Primary mucosal melanomas (MMs) are uncommon tumors originating from melanocytes located in the mucous membranes at various anatomic sites within the body. MM significantly differs from cutaneous melanoma (CM) regarding epidemiology, genetic profile, clinical presentation, and response to therapies. Despite these differences, that have important implications for both disease diagnosis and prognosis, MMs are usually treated in the same way as CM but exhibit a lower response rate to immunotherapy leading to a poorer survival rate. Furthermore, a high inter-patient variability can be observed in relation to therapeutic response. Recently, novel "omics" techniques have evidenced that MM lesions have different genomic, molecular, and metabolic landscapes as compared with CM lesions, thus explaining the heterogeneity of the response. Such specific molecular aspects might be useful to identify new biomarkers aimed at improving the diagnosis and selection of MM patients who could benefit from immunotherapy or targeted therapy. In this review, we have focused on relevant molecular and clinical advancements for the different MM subtypes in order to describe the updated knowledge relating to main diagnostic, clinical, and therapeutic implications as well as to provide hints on likely future directions.
Collapse
|
137
|
Nelson BE, Roszik J, Janku F, Hong DS, Kato S, Naing A, Piha-Paul S, Fu S, Tsimberidou A, Cabanillas M, Busaidy NL, Javle M, Byers LA, Heymach JV, Meric-Bernstam F, Subbiah V. BRAF v600E-mutant cancers treated with vemurafenib alone or in combination with everolimus, sorafenib, or crizotinib or with paclitaxel and carboplatin (VEM-PLUS) study. NPJ Precis Oncol 2023; 7:19. [PMID: 36801912 PMCID: PMC9938883 DOI: 10.1038/s41698-022-00341-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 12/14/2022] [Indexed: 02/20/2023] Open
Abstract
Combined BRAF + MEK inhibition is FDA approved for BRAF V600E-mutant solid tumors except for colorectal cancer. However, beyond MAPK mediated resistance several other mechanisms of resistance such as activation of CRAF, ARAF, MET, P13K/AKT/mTOR pathway exist among other complex pathways. In the VEM-PLUS study, we performed a pooled analysis of four phase one studies evaluating the safety and efficacy of vemurafenib monotherapy and vemurafenib combined with targeted therapies (sorafenib, crizotinib, or everolimus) or carboplatin plus paclitaxel in advanced solid tumors harboring BRAF V600 mutations. When vemurafenib monotherapy was compared with the combination regimens, no significant differences in OS or PFS durations were noted, except for inferior OS in the vemurafenib and paclitaxel and carboplatin trial (P = 0.011; HR, 2.4; 95% CI, 1.22-4.7) and in crossover patients (P = 0.0025; HR, 2.089; 95% CI, 1.2-3.4). Patients naïve to prior BRAF inhibitors had statistically significantly improved OS at 12.6 months compared to 10.4 months in the BRAF therapy refractory group (P = 0.024; HR, 1.69; 95% CI 1.07-2.68). The median PFS was statistically significant between both groups, with 7 months in the BRAF therapy naïve group compared to 4.7 months in the BRAF therapy refractory group (P = 0.016; HR, 1.80; 95% CI 1.11-2.91). The confirmed ORR in the vemurafenib monotherapy trial (28%) was higher than that in the combination trials. Our findings suggest that, compared with vemurafenib monotherapy, combinations of vemurafenib with cytotoxic chemotherapy or with RAF- or mTOR-targeting agents do not significantly extend the OS or PFS of patients who have solid tumors with BRAF V600E mutations. Gaining a better understanding of the molecular mechanisms of BRAF inhibitor resistance, balancing toxicity and efficacy with novel trial designs are warranted.
Collapse
Affiliation(s)
- Blessie Elizabeth Nelson
- grid.240145.60000 0001 2291 4776Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Jason Roszik
- grid.240145.60000 0001 2291 4776Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Filip Janku
- grid.240145.60000 0001 2291 4776Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - David S. Hong
- grid.240145.60000 0001 2291 4776Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Shumei Kato
- grid.240145.60000 0001 2291 4776Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Aung Naing
- grid.240145.60000 0001 2291 4776Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Sarina Piha-Paul
- grid.240145.60000 0001 2291 4776Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Siqing Fu
- grid.240145.60000 0001 2291 4776Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Apostolia Tsimberidou
- grid.240145.60000 0001 2291 4776Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Maria Cabanillas
- grid.240145.60000 0001 2291 4776Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Naifa Lamki Busaidy
- grid.240145.60000 0001 2291 4776Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Milind Javle
- grid.240145.60000 0001 2291 4776Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Lauren Averett Byers
- grid.240145.60000 0001 2291 4776Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - John V. Heymach
- grid.240145.60000 0001 2291 4776Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Funda Meric-Bernstam
- grid.240145.60000 0001 2291 4776Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Vivek Subbiah
- Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
138
|
Allard-Coutu A, Dobson V, Schmitz E, Shah H, Nessim C. The Evolution of the Sentinel Node Biopsy in Melanoma. Life (Basel) 2023; 13:life13020489. [PMID: 36836846 PMCID: PMC9966203 DOI: 10.3390/life13020489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/11/2023] [Accepted: 02/02/2023] [Indexed: 02/12/2023] Open
Abstract
The growing repertoire of approved immune-checkpoint inhibitors and targeted therapy has revolutionized the adjuvant treatment of melanoma. While the treatment of primary cutaneous melanoma remains wide local excision (WLE), the management of regional lymph nodes continues to evolve in light of practice-changing clinical trials and dramatically improved adjuvant therapy. With large multicenter studies reporting no benefit in overall survival for completion lymph node dissection (CLND) after a positive sentinel node biopsy (SLNB), controversy remains regarding patient selection and clinical decision-making. This review explores the evolution of the SLNB in cutaneous melanoma in the context of a rapidly changing adjuvant treatment landscape, summarizing the key clinical trials which shaped current practice guidelines.
Collapse
Affiliation(s)
- Alexandra Allard-Coutu
- Department of General Surgery, University of Ottawa, Ottawa, ON K1N 6N5, Canada
- Correspondence:
| | | | - Erika Schmitz
- Department of General Surgery, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Hely Shah
- Department of Medical Oncology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Carolyn Nessim
- Department of General Surgery, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
139
|
Development and validation of a decision model for the evaluation of novel lung cancer treatments in the Netherlands. Sci Rep 2023; 13:2349. [PMID: 36759641 PMCID: PMC9911639 DOI: 10.1038/s41598-023-29286-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/01/2023] [Indexed: 02/11/2023] Open
Abstract
Recent discoveries in molecular diagnostics and drug treatments have improved the treatment of patients with advanced (inoperable) non-squamous non-small cell lung cancer (NSCLC) from solely platinum-based chemotherapy to more personalized treatment, including targeted therapies and immunotherapies. However, these improvements come at considerable costs, highlighting the need to assess their cost-effectiveness in order to optimize lung cancer care. Traditionally, cost-effectiveness models for the evaluation of new lung cancer treatments were based on the findings of the randomized control trials (RCTs). However, the strict RCT inclusion criteria make RCT patients not representative of patients in the real-world. Patients in RCTs have a better prognosis than patients in a real-world setting. Therefore, in this study, we developed and validated a diagnosis-treatment decision model for patients with advanced (inoperable) non-squamous NSCLC based on real-world data in the Netherlands. The model is a patient-level microsimulation model implemented as discrete event simulation with five health events. Patients are simulated from diagnosis to death, including at most three treatment lines. The base-model (non-personalized strategy) was populated using real-world data of patients treated with platinum-based chemotherapy between 2008 and 2014 in one of six Dutch teaching hospitals. To simulate personalized care, molecular tumor characteristics were incorporated in the model based on the literature. The impact of novel targeted treatments and immunotherapies was included based on published RCTs. To validate the model, we compared survival under a personalized treatment strategy with observed real-world survival. This model can be used for health-care evaluation of personalized treatment for patients with advanced (inoperable) NSCLC in the Netherlands.
Collapse
|
140
|
Pala L, De Pas T, Pagan E, Minucci S, Catania C, Digiacomo N, Cocorocchio E, Laszlo D, Di Muzio A, Barigazzi C, Stucchi E, De Grandi L, Stucchi S, Viale G, Gelber RD, Bagnardi V, Conforti F. Improved outcomes in women with BRAF-mutant melanoma treated with BRAF/MEK-targeted therapy across randomized clinical trials. A systematic review and meta-analysis. Semin Oncol 2023; 50:34-39. [PMID: 36967333 DOI: 10.1053/j.seminoncol.2023.03.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 03/12/2023] [Accepted: 03/14/2023] [Indexed: 06/05/2023]
Abstract
Available evidence suggests that in patients with advanced BRAF V600-mutant melanoma treated with the combination of BRAF and MEK inhibitors, gender could be associated with survival outcome. We performed a systematic review and meta-analysis of all randomized clinical trials (RCTs) testing the combination of BRAF and MEK inhibitors, to assess the interaction between treatment effect and patients' gender. We searched PubMed, MEDLINE, Embase, and Scopus, for phase II and III RCTs up to January 30, 2022. We included all RCTs that enrolled patients with BRAF V600-mutant advanced cutaneous melanoma and assessed combinations of BRAF and MEK inhibitors versus BRAF inhibitor monotherapy. Our aim was to assess differences if any in treatment efficacy between men and women, measured in terms of the differences in progression-free survival (PFS) and overall survival (OS) log-hazard ratios (log-HRs). We calculated the pooled PFS- and OS-HRs with 95% confidence intervals (CIs) in men and women using a random-effects model and assessed the heterogeneity between the estimates using an interaction test. Five RCTs that enrolled a total of 2,113 patients were included in the analysis. In women, the combination of BRAF and MEK inhibitors halved the risk of progression or death as compared with BRAF inhibitor monotherapy with a pooled PFS-HR of 0.50 (95%CI 0.41-0.61). In men, the benefit obtained with BRAF and MEK inhibitors was smaller with a pooled PFS-HR of 0.63 (95%CI 0.54-0.74), P-heterogeneity = .05. A similar trend was observed for OS where the pooled OS-HR was 0.62 (95%CI 0.48-0.80) in women and only 0.78, (95%CI 0.67-0.92) in men, P-heterogeneity = 0.11. These results support meaningful gender-based heterogeneity of response to combination of BRAF and MEK inhibitors targeted therapy in patients with advanced BRAF-mutant melanoma, that should be considered in future research to improve treatment effectiveness.
Collapse
Affiliation(s)
- Laura Pala
- Medical Oncology Unit, Humanitas Gavazzeni, Bergamo, Italy.
| | - Tommaso De Pas
- Medical Oncology Unit, Humanitas Gavazzeni, Bergamo, Italy
| | - Eleonora Pagan
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | - Saverio Minucci
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Chiara Catania
- Medical Oncology Unit, Humanitas Gavazzeni, Bergamo, Italy
| | | | | | - Daniele Laszlo
- Medical Oncology Unit, Humanitas Gavazzeni, Bergamo, Italy
| | - Antonio Di Muzio
- Department of Biomedical Sciences, Humanitas University, Rozzano, Italy
| | - Chiara Barigazzi
- Medical Oncology Unit, Humanitas Gavazzeni, Bergamo, Italy; Department of Biomedical Sciences, Humanitas University, Rozzano, Italy
| | - Erika Stucchi
- Medical Oncology Unit, Humanitas Gavazzeni, Bergamo, Italy; Department of Biomedical Sciences, Humanitas University, Rozzano, Italy
| | - Laura De Grandi
- Medical Oncology Unit, Humanitas Gavazzeni, Bergamo, Italy; Department of Biomedical Sciences, Humanitas University, Rozzano, Italy
| | - Sara Stucchi
- Medical Oncology Unit, Humanitas Gavazzeni, Bergamo, Italy
| | - Giuseppe Viale
- Department of Pathology, European Institute of Oncology & University of Milan, Milan, Italy
| | - Richard D Gelber
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Harvard T.H. Chan School of Public Health and Frontier Science & Technology, Research Foundation, Boston, MA, USA
| | - Vincenzo Bagnardi
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, Milan, Italy
| | - Fabio Conforti
- Medical Oncology Unit, Humanitas Gavazzeni, Bergamo, Italy
| |
Collapse
|
141
|
Vasudevan HN, Susko MS, Ma L, Nakamura JL, Raleigh DR, Boreta L, Fogh S, Theodosopoulos PV, McDermott MW, Tsai KK, Sneed PK, Braunstein SE. Mutational Status and Clinical Outcomes Following Systemic Therapy with or without Focal Radiation for Resected Melanoma Brain Metastases. World Neurosurg 2023; 170:e514-e519. [PMID: 36400359 DOI: 10.1016/j.wneu.2022.11.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/12/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Brain metastases occur frequently in advanced melanoma and traditionally require surgery and radiation therapy. New evidence demonstrates that systemic therapies are effective for controlling metastatic melanoma brain metastases. This study evaluated outcomes after resection of melanoma brain metastases treated with systemic therapy, with or without focal radiotherapy. METHODS All patients received immunotherapy or BRAF/MEK inhibitors preoperatively or in the immediate 3 months postoperatively. Resection cavity failure, distant central nervous system progression, and adverse radiation effects were reported in the presence and absence of focal radiotherapy using the Kaplan-Meier method. RESULTS Between 2011 and 2020, 37 resection cavities in 29 patients met criteria for analysis. Of lesions, 22 (59%) were treated with focal radiotherapy, and 15 (41%) were treated with targeted therapy or immunotherapy alone. The 12- and 24-month freedom from local recurrence was 64.8% (95% confidence interval [CI] 42.1%-99.8%) and 46.3% (95% CI 24.5%-87.5%), respectively, for systemic therapy alone and 93.3% (95% CI 81.5%-100%) at both time points for focal radiotherapy (P = 0.01). On univariate analysis, focal radiotherapy was the only significant factor associated with reduction of local recurrence risk (hazard ratio 0.10, 95% CI 0.01-0.85; P = 0.04). There were no significant differences in central nervous system progression-free survival or overall survival between patients who received systemic therapy plus focal radiotherapy compared with systemic therapy alone. BRAF mutation status was reviewed for either the brain metastasis (n = 9 patients, 31%) or the primary site (n = 20 patients, 69%), and patients harboring BRAFV600E mutations had worse progression-free survival (P = 0.043). CONCLUSIONS Focal radiotherapy with systemic therapy for resected melanoma brain metastases significantly decreased resection cavity recurrence compared with systemic therapy alone. BRAF mutation status correlated with poorer outcomes.
Collapse
Affiliation(s)
- Harish N Vasudevan
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - Matthew S Susko
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - Lijun Ma
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - Jean L Nakamura
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - David R Raleigh
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA; Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Lauren Boreta
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - Shannon Fogh
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - Philip V Theodosopoulos
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - Michael W McDermott
- Miami Neuroscience Institute, Baptist Health South Florida, Miami, Florida, USA
| | - Katy K Tsai
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Penny K Sneed
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - Steve E Braunstein
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA.
| |
Collapse
|
142
|
Dodoo GN, De B, Lee SS, Abi Jaoude J, Vauthey JN, Tzeng CWD, Tran Cao HS, Katlowitz KA, Mandel JJ, Beckham TH, Minsky BD, Smith GL, Holliday EB, Koong AC, Das P, Taniguchi CM, Javle M, Koay EJ, Ludmir EB. Brain Metastases from Biliary Tract Cancer: Case Series and Clinicogenomic Analysis. Oncologist 2023; 28:327-332. [PMID: 36715178 PMCID: PMC10078902 DOI: 10.1093/oncolo/oyac273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 11/17/2022] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Limited data from small series have suggested that brain metastases from biliary tract cancers (BrM-BTC) affect ≤2% of patients with BTC. We sought to review our experience with patients with BrM-BTC and to identify associations of tumor-related molecular alterations with outcomes. MATERIALS AND METHODS A retrospective review of patients with BTC seen at a tertiary referral center from 2005 to 2021 was performed; patients with BrM-BTC were identified, and clinical and molecular data were collected. RESULTS Twenty-one of 823 patients with BTC (2.6%) developed BrM. For patients with BrM-BTC, median follow-up time was 27.9 months after primary BTC diagnosis and 3.1 months after BrM diagnosis. Median time from primary diagnosis to diagnosis of BrM was 14.4 [range, 1.1-66.0] months. Median overall survival (OS) from primary diagnosis was 31.5 [2.9-99.8] months and median OS from BrM diagnosis was 4.2 [0.2-33.8] months. Patients who underwent BrM-directed therapy trended toward longer OS following BrM diagnosis than patients receiving supportive care only (median 6.5 vs 0.8 months, P = .060). The BrM-BTC cohort was enriched for BRAF (30%), PIK3CA (25%), and GNAS (20%) mutations. patients with BrM-BTC with BRAF mutations trended toward longer OS following BrM diagnosis (median 13.1 vs 4.2 months, P = .131). CONCLUSION This is the largest series of patients with BrM-BTC to date and provides molecular characterization of this rare subgroup of patients with BTC. Patients with BrM-BTC may be more likely to have BRAF mutations. With advances in targeted therapy for patients with BTC with actionable mutations, continued examination of shifting patterns of failure, with emphasis on BrM, is warranted.
Collapse
Affiliation(s)
- Grace N Dodoo
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brian De
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sunyoung S Lee
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph Abi Jaoude
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jean-Nicolas Vauthey
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ching-Wei D Tzeng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hop S Tran Cao
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kalman A Katlowitz
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA.,Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jacob J Mandel
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Thomas H Beckham
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bruce D Minsky
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Grace L Smith
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Emma B Holliday
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Albert C Koong
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Prajnan Das
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cullen M Taniguchi
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Milind Javle
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eugene J Koay
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ethan B Ludmir
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
143
|
Bouffet E, Geoerger B, Moertel C, Whitlock JA, Aerts I, Hargrave D, Osterloh L, Tan E, Choi J, Russo M, Fox E. Efficacy and Safety of Trametinib Monotherapy or in Combination With Dabrafenib in Pediatric BRAF V600-Mutant Low-Grade Glioma. J Clin Oncol 2023; 41:664-674. [PMID: 36375115 PMCID: PMC9870224 DOI: 10.1200/jco.22.01000] [Citation(s) in RCA: 94] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/09/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
PURPOSE BRAF V600 mutations occur in many childhood cancers, including approximately 20% of low-grade gliomas (LGGs). Here, we describe a phase I/II study establishing pediatric dosing and pharmacokinetics of trametinib with or without dabrafenib, as well as efficacy and safety in a disease-specific cohort with BRAF V600-mutant LGG; other cohorts will be reported elsewhere. METHODS This is a four-part, phase I/II study (ClinicalTrials.gov identifier: NCT02124772) in patients age < 18 years with relapsed/refractory malignancies: trametinib monotherapy dose finding (part A) and disease-specific expansion (part B), and dabrafenib + trametinib dose finding (part C) and disease-specific expansion (part D). The primary objective assessed in all patients in parts A and C was to determine pediatric dosing on the basis of steady-state pharmacokinetics. Disease-specific efficacy and safety (across parts A-D) were secondary objectives. RESULTS Overall, 139 patients received trametinib (n = 91) or dabrafenib + trametinib (n = 48). Trametinib dose-limiting toxicities in > 1 patient (part A) included mucosal inflammation (n = 3) and hyponatremia (n = 2). There were no dose-limiting toxicities with combination therapy (part C). The recommended phase II dose of trametinib, with or without dabrafenib, was 0.032 mg/kg once daily for patients age < 6 years and 0.025 mg/kg once daily for patients age ≥ 6 years; dabrafenib dosing in the combination was as previously identified for monotherapy. In 49 patients with BRAF V600-mutant glioma (LGG, n = 47) across all four study parts, independently assessed objective response rates were 15% (95% CI, 1.9 to 45.4) for monotherapy (n = 13) and 25% (95% CI, 12.1 to 42.2) for combination (n = 36). Adverse event-related treatment discontinuations were more common with monotherapy (54% v 22%). CONCLUSION The trial design provided efficient evaluation of pediatric dosing, safety, and efficacy of single-agent and combination targeted therapy. Age-based and weight-based dosing of trametinib with or without dabrafenib achieved target concentrations with manageable safety and demonstrated clinical efficacy and tolerability in BRAF V600-mutant LGG.
Collapse
Affiliation(s)
- Eric Bouffet
- Department of Paediatrics, The Hospital for Sick Children/University of Toronto, Toronto, ON, Canada
| | - Birgit Geoerger
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Center, INSERM U1015, Université Paris-Saclay, Villejuif, France
| | | | - James A Whitlock
- Department of Paediatrics, The Hospital for Sick Children/University of Toronto, Toronto, ON, Canada
| | - Isabelle Aerts
- Institut Curie, PSL Research University, Oncology Center SIREDO, Paris, France
| | - Darren Hargrave
- Great Ormond Street Hospital for Children, London, United Kingdom
| | | | - Eugene Tan
- Novartis Pharmaceuticals Corporation, East Hanover, NJ
| | - Jeea Choi
- Novartis Pharmaceuticals Corporation, East Hanover, NJ
| | - Mark Russo
- Novartis Pharmaceuticals Corporation, East Hanover, NJ
| | - Elizabeth Fox
- Comprehensive Cancer Center, St Jude Children's Research Hospital, Memphis, TN
| |
Collapse
|
144
|
Naik RR, Shakya AK. Exploring the chemotherapeutic potential of currently used kinase inhibitors: An update. Front Pharmacol 2023; 13:1064472. [PMID: 36699049 PMCID: PMC9868582 DOI: 10.3389/fphar.2022.1064472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 11/28/2022] [Indexed: 01/11/2023] Open
Abstract
Protein kinases are enzymes that transfer phosphate to protein, resulting in the modification of the protein. The human genome encodes approximately 538 kinases. Kinases play a role in maintaining a number of cellular processes, including control of the cell cycle, metabolism, survival, and differentiation. Protein kinase dysregulation causes several diseases, and it has been shown that numerous kinases are deregulated in cancer. The oncogenic potential of these kinases is increased by a number of processes, including overexpression, relocation, fusion point mutations, and the disruption of upstream signaling. Understanding of the mechanism or role played by kinases has led to the development of a large number of kinase inhibitors with promising clinical benefits. In this review, we discuss FDA-approved kinase inhibitors and their mechanism, clinical benefits, and side effects, as well as the challenges of overcoming some of their side effects and future prospects for new kinase inhibitor discovery.
Collapse
Affiliation(s)
- Rajashri R. Naik
- Faculty of Allied Medical Sciences, Pharmacological and Diagnostic Research Center, Al-Ahliyya Amman University, Amman, Jordan
| | - Ashok K. Shakya
- Faculty of Pharmacy, Pharmacological and Diagnostic Research Center, Al-Ahliyya Amman University, Amman, Jordan,*Correspondence: Ashok K. Shakya,
| |
Collapse
|
145
|
Chen D, Su X, Zhu L, Jia H, Han B, Chen H, Liang Q, Hu C, Yang H, Liu L, Li P, Wei W, Zhao Y. Papillary thyroid cancer organoids harboring BRAF V600E mutation reveal potentially beneficial effects of BRAF inhibitor-based combination therapies. J Transl Med 2023; 21:9. [PMID: 36624452 PMCID: PMC9827684 DOI: 10.1186/s12967-022-03848-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 12/23/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUNDS Papillary thyroid cancer (PTC), which is often driven by acquired somatic mutations in BRAF genes, is the most common pathologic type of thyroid cancer. PTC has an excellent prognosis after treatment with conventional therapies such as surgical resection, thyroid hormone therapy and adjuvant radioactive iodine therapy. Unfortunately, about 20% of patients develop regional recurrence or distant metastasis, making targeted therapeutics an important treatment option. Current in vitro PTC models are limited in representing the cellular and mutational characteristics of parental tumors. A clinically relevant tool that predicts the efficacy of therapy for individuals is urgently needed. METHODS Surgically removed PTC tissue samples were dissociated, plated into Matrigel, and cultured to generate organoids. PTC organoids were subsequently subjected to histological analysis, DNA sequencing, and drug sensitivity assays, respectively. RESULTS We established 9 patient-derived PTC organoid models, 5 of which harbor BRAFV600E mutation. These organoids have been cultured stably for more than 3 months and closely recapitulated the histological architectures as well as mutational landscapes of the respective primary tumors. Drug sensitivity assays of PTC organoid cultures demonstrated the intra- and inter-patient specific drug responses. BRAFV600E inhibitors, vemurafenib and dabrafenib monotherapy was mildly effective in treating BRAFV600E-mutant PTC organoids. Nevertheless, BRAF inhibitors in combination with MEK inhibitors, RTK inhibitors, or chemotherapeutic agents demonstrated improved efficacy compared to BRAF inhibition alone. CONCLUSIONS These data indicate that patient-derived PTC organoids may be a powerful research tool to investigate tumor biology and drug responsiveness, thus being useful to validate or discover targeted drug combinations.
Collapse
Affiliation(s)
- Dong Chen
- grid.440601.70000 0004 1798 0578Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036 China
| | - Xi Su
- grid.440601.70000 0004 1798 0578Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036 China
| | - Lizhang Zhu
- grid.440601.70000 0004 1798 0578Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036 China
| | - Hao Jia
- grid.440601.70000 0004 1798 0578Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036 China
| | - Bin Han
- grid.440601.70000 0004 1798 0578Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036 China
| | - Haibo Chen
- grid.440601.70000 0004 1798 0578Department of Nuclear Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036 China
| | - Qingzhuang Liang
- grid.440601.70000 0004 1798 0578Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036 China
| | - Chenchen Hu
- grid.440601.70000 0004 1798 0578Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036 China
| | - Hao Yang
- grid.440601.70000 0004 1798 0578Department of Nuclear Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036 China
| | - Lisa Liu
- grid.264727.20000 0001 2248 3398Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19122 USA
| | - Peng Li
- grid.440601.70000 0004 1798 0578Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036 China
| | - Wei Wei
- grid.440601.70000 0004 1798 0578Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036 China
| | - Yongsheng Zhao
- grid.440601.70000 0004 1798 0578Department of Thyroid and Breast Surgery, Peking University Shenzhen Hospital, Shenzhen, 518036 China ,grid.440601.70000 0004 1798 0578Department of Nuclear Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036 China
| |
Collapse
|
146
|
Guaitoli G, Zullo L, Tiseo M, Dankner M, Rose AAN, Facchinetti F. Non-small-cell lung cancer: how to manage BRAF-mutated disease. Drugs Context 2023; 12:dic-2022-11-3. [PMID: 37168877 PMCID: PMC10166262 DOI: 10.7573/dic.2022-11-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 03/17/2023] [Indexed: 05/13/2023] Open
Abstract
BRAF mutations are reported in about 3-5% of non-small-cell lung cancer (NSCLC), almost exclusively in adenocarcinoma histology, and are classified into three different classes. The segmentation of BRAF mutations into V600 (class 1) and non-V600 (classes 2 and 3) relies on their biological characteristics and is of interest for predicting the therapeutic benefit of targeted therapies and immunotherapy. Given the relative rarity of this molecular subset of disease, evidence supporting treatment choices is limited. This review aims to offer a comprehensive update about available therapeutic options for patients with NSCLC harbouring BRAF mutations to guide the physician in the choice of treatment strategies. We collected the most relevant available data, from single-arm phase II studies and retrospective analyses conducted in advanced NSCLC, regarding the efficacy of BRAF and MEK inhibitors in both V600 and non-V600 BRAF mutations. We included case reports and smaller experiences that could provide information on specific alterations. With respect to immunotherapy, we reviewed retrospective evidence on immune-checkpoint inhibitors in this molecular subset, whereas data about chemo-immunotherapy in this molecular subgroup are lacking. Moreover, we included the available, though limited, retrospective evidence of immunotherapy as consolidation after chemo-radiation for unresectable stage III BRAF-mutant NSCLC, and an overview of ongoing clinical trials in the peri-operative setting that could open new perspectives in the future.
Collapse
Affiliation(s)
- Giorgia Guaitoli
- Université Paris-Saclay, Gustave Roussy, INSERM U981, Villejuif, France
- PhD Program Clinical & Experimental Medicine, University of Modena & Reggio Emilia, Modena, Italy
| | - Lodovica Zullo
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
- Department of Cancer Medicine, Gustave Roussy Cancer Campus, Villejuif, France
| | - Marcello Tiseo
- Department of Medicine and Surgery, University Hospital of Parma, Parma, Italy
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Matthew Dankner
- Lady Davis Institute, Segal Cancer Centre, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, Québec, Canada
- Faculty of Medicine, McGill University, Montréal, Québec, Canada
| | - April AN Rose
- Lady Davis Institute, Segal Cancer Centre, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Department of Oncology, McGill University, Montréal, Québec, Canada
| | - Francesco Facchinetti
- Université Paris-Saclay, Gustave Roussy, INSERM U981, Villejuif, France
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
147
|
Garutti M, Bergnach M, Polesel J, Palmero L, Pizzichetta MA, Puglisi F. BRAF and MEK Inhibitors and Their Toxicities: A Meta-Analysis. Cancers (Basel) 2022; 15:cancers15010141. [PMID: 36612138 PMCID: PMC9818023 DOI: 10.3390/cancers15010141] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/15/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
PURPOSE This meta-analysis summarizes the incidence of treatment-related adverse events (AE) of BRAFi and MEKi. METHODS A systematic search of Medline/PubMed was conducted to identify suitable articles published in English up to 31 December 2021. The primary outcomes were profiles for all-grade and grade 3 or higher treatment-related AEs, and the analysis of single side effects belonging to both categories. RESULTS The overall incidence of treatment-related all-grade Aes was 99% for Encorafenib (95% CI: 0.97-1.00) and 97% for Trametinib (95% CI: 0.92-0.99; I2 = 66%) and Binimetinib (95% CI: 0.94-0.99; I2 = 0%). In combined therapies, the rate was 98% for both Vemurafenib + Cobimetinib (95% CI: 0.96-0.99; I2 = 77%) and Encorafenib + Binimetinib (95% CI: 0.96-1.00). Grade 3 or higher adverse events were reported in 69% of cases for Binimetinib (95% CI: 0.50-0.84; I2 = 71%), 68% for Encorafenib (95% CI: 0.61-0.74), and 72% for Vemurafenib + Cobimetinib (95% CI: 0.65-0.79; I2 = 84%). The most common grade 1-2 AEs were pyrexia (43%) and fatigue (28%) for Dabrafenib + Trametinib and diarrhea for both Vemurafenib + Cobimetinib (52%) and Encorafenib + Binimetinib (34%). The most common AEs of grade 3 or higher were pyrexia, rash, and hypertension for Dabrafenib + Trametinib (6%), rash and hypertension for Encorafenib + Binimetinib (6%), and increased AST and ALT for Vemurafenib + Cobimetinib (10%). CONCLUSIONS Our study provides comprehensive data on treatment-related adverse events of BRAFi and MEKi combination therapies, showing related toxicity profiles to offer a helpful tool for clinicians in the choice of therapy.
Collapse
Affiliation(s)
- Mattia Garutti
- CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
- Correspondence: ; Tel.: +39-04-3465-9092
| | | | - Jerry Polesel
- Unit of Cancer Epidemiology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Lorenza Palmero
- CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Maria Antonietta Pizzichetta
- CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
- Department of Dermatology, University of Trieste, 34123 Trieste, Italy
| | - Fabio Puglisi
- CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
- Department of Medicine, University of Udine, 33100 Udine, Italy
| |
Collapse
|
148
|
Ohshima S, Ueki Y, Yokoyama Y, Takahashi T, Shodo R, Yamazaki K, Okabe R, Matsuyama H, Togashi T, Takatsuka S, Takenouchi T, Horii A. Treatment outcomes of mucosal melanoma of head and neck: Efficacy of immune checkpoint inhibitors for advanced disease. Front Surg 2022; 9:1032626. [PMID: 37082097 PMCID: PMC10112385 DOI: 10.3389/fsurg.2022.1032626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/12/2022] [Indexed: 12/27/2022] Open
Abstract
BackgroundHead and neck mucosal melanoma (HNMM) is a rare and aggressive subtype of melanoma. HNMM often develops as a recurrent or metastatic disease, and its prognosis is worse than that of cutaneous melanoma. Recent large-scale clinical studies have reported favorable outcomes with immune checkpoint inhibitors (ICIs) for melanoma. However, these clinical trials included only a small number of HNMM cases. This study aimed to estimate treatment outcomes and prognostic predictors of ICIs for advanced HNMM.MethodsCases of advanced HNMM, defined as unresectable or metastatic HNMM at the initial diagnosis (five patients) or development of recurrent/metastatic HNMM after initial treatment (27 patients), were included in this study. Survival analysis and a search for prognostic factors were performed for these 32 patients. Furthermore, the detailed clinical course of patients who received ICI treatment was investigated.ResultsThe median overall survival (OS) of 32 patients with advanced HNMM was 25.3 months. The estimated 1-, 3-, and 5-year OS rates were 68.4%, 42.8%, and 34.3%, respectively. Fourteen patients (43.7%) received ICIs, whereas 18 (56.3%) did not. Univariate analysis showed that ICI treatment was the only factor associated with a better 1-year OS. Patients who received ICI treatment had significantly longer OS (median OS: not reached, 1-year OS: 85.7%) than those who did not (median OS: 11.3 months, 1-year OS: 54.5%). The overall response and disease control rates of patients who received ICI treatment were 50% and 64.3%, respectively. Patients who achieved complete response (CR) or partial response (PR) to ICI treatment survived significantly longer (1-year OS: 100%) than those who did not (1-year OS: 71.4%). Among the five patients who discontinued ICI treatment due to severe immune-related adverse events (irAEs), four did not receive salvage treatments but showed durable treatment effects and survived for 9.8–54.2 months at the end of the follow-up period.ConclusionsICI treatment achieved a favorable OS for advanced HNMM. CR/PR to ICI treatment and discontinuation owing to severe irAEs were favorable predictors of OS.
Collapse
Affiliation(s)
- Shusuke Ohshima
- Department of Otolaryngology Head and Neck Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yushi Ueki
- Department of Otolaryngology Head and Neck Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Correspondence: Yushi Ueki
| | - Yusuke Yokoyama
- Department of Otolaryngology Head and Neck Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Takeshi Takahashi
- Department of Otolaryngology Head and Neck Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ryusuke Shodo
- Department of Otolaryngology Head and Neck Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Keisuke Yamazaki
- Department of Otolaryngology Head and Neck Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ryuichi Okabe
- Division of Otorhinolaryngology, Nagaoka Red Cross Hospital, Niigata, Japan
| | - Hiroshi Matsuyama
- Division of Otorhinolaryngology, Niigata City General Hospital, Niigata, Japan
| | - Takafumi Togashi
- Division of Head and Neck Surgery, Niigata Cancer Center Hospital, Niigata, Japan
| | - Sumiko Takatsuka
- Division of Dermatology, Niigata Cancer Center Hospital, Niigata, Japan
| | | | - Arata Horii
- Department of Otolaryngology Head and Neck Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
149
|
Seyhan AA, Carini C. Insights and Strategies of Melanoma Immunotherapy: Predictive Biomarkers of Response and Resistance and Strategies to Improve Response Rates. Int J Mol Sci 2022; 24:ijms24010041. [PMID: 36613491 PMCID: PMC9820306 DOI: 10.3390/ijms24010041] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/10/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Despite the recent successes and durable responses with immune checkpoint inhibitors (ICI), many cancer patients, including those with melanoma, do not derive long-term benefits from ICI therapies. The lack of predictive biomarkers to stratify patients to targeted treatments has been the driver of primary treatment failure and represents an unmet medical need in melanoma and other cancers. Understanding genomic correlations with response and resistance to ICI will enhance cancer patients' benefits. Building on insights into interplay with the complex tumor microenvironment (TME), the ultimate goal should be assessing how the tumor 'instructs' the local immune system to create its privileged niche with a focus on genomic reprogramming within the TME. It is hypothesized that this genomic reprogramming determines the response to ICI. Furthermore, emerging genomic signatures of ICI response, including those related to neoantigens, antigen presentation, DNA repair, and oncogenic pathways, are gaining momentum. In addition, emerging data suggest a role for checkpoint regulators, T cell functionality, chromatin modifiers, and copy-number alterations in mediating the selective response to ICI. As such, efforts to contextualize genomic correlations with response into a more insightful understanding of tumor immune biology will help the development of novel biomarkers and therapeutic strategies to overcome ICI resistance.
Collapse
Affiliation(s)
- Attila A. Seyhan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI 02912, USA
- Legorreta Cancer Center, Brown University, Providence, RI 02912, USA
- Correspondence:
| | - Claudio Carini
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, New Hunt’s House, Guy’s Campus, King’s College London, London SE1 1UL, UK
- Biomarkers Consortium, Foundation of the National Institute of Health, Bethesda, MD 20892, USA
| |
Collapse
|
150
|
Eriksen M, Pfeiffer P, Rohrberg KS, Yde CW, Petersen LN, Poulsen LØ, Qvortrup C. A phase II study of daily encorafenib in combination with biweekly cetuximab in patients with BRAF V600E mutated metastatic colorectal cancer: the NEW BEACON study. BMC Cancer 2022; 22:1321. [PMID: 36527039 PMCID: PMC9758813 DOI: 10.1186/s12885-022-10420-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Patients with BRAF V600E mutated metastatic colorectal cancer (mCRC) have a poor prognosis. The introduction of BRAF targeted therapy with encorafenib and weekly administered cetuximab have shown improved survival with a median progression free survival (PFS) of 4.3 months. However, a regimen with cetuximab given every second week may have comparable efficacy and is more convenient for patients. While BRAF targeted therapy is a new standard therapy in pre-treated patients with BRAF V600E mutated mCRC, resistance invariably occurs and is an emerging challenge. The aim of this study is to investigate the efficacy and tolerability of cetuximab given every second week in combination with daily encorafenib and to explore the correlation between markers of resistance and outcome. METHODS The study is an open label, single arm, phase II study, investigating the efficacy and tolerability of cetuximab given every second week in combination with encorafenib in patients with BRAF V600E mutated mCRC. Furthermore, we will be investigating mechanisms of response and resistance against BRAF targeted therapy though comprehensive genomic profiling on tumor tissue and blood for circulating tumor DNA analysis. A total of 53 patients (19 + 34 in two steps) will be included according to Simon's optimal two stage design. The primary end point of the study is 2 months PFS rate. DISCUSSION By combining BRAF inhibitor with cetuximab given every second week we can halve the number of visits in the hospital compared to the currently approved regimen with weekly cetuximab. This seems particularly relevant in a group of patients with a median overall survival of 9.3 months. Resistance after initial response to targeted therapy can be either adaptive (e.g., epigenetic, or transcriptomic alterations) or acquired (selective genetic alterations - e.g., activating de novo mutations) resistance. It is of great importance to untangle these complex mechanisms of resistance in patients with BRAF V600E mutated mCRC to improve treatment strategies in the future potentially even further. TRIAL REGISTRATION EU Clinical Trial Register, Eudract no. 2020-003283-10 . Registered on 11 November 2020.
Collapse
Affiliation(s)
- Martina Eriksen
- grid.475435.4Department of Oncology, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark ,grid.5254.60000 0001 0674 042XFaculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Per Pfeiffer
- grid.7143.10000 0004 0512 5013Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Kristoffer Staal Rohrberg
- grid.475435.4Department of Oncology, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Christina Westmose Yde
- grid.475435.4Department of Genomic Medicine, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Lone Nørgård Petersen
- grid.475435.4Department of Oncology, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | | | - Camilla Qvortrup
- grid.475435.4Department of Oncology, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark ,grid.5254.60000 0001 0674 042XFaculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|