601
|
Karlsson MJ, Costa Svedman F, Tebani A, Kotol D, Höiom V, Fagerberg L, Edfors F, Uhlén M, Egyhazi Brage S, Maddalo G. Inflammation and Apolipoproteins Are Potential Biomarkers for Stratification of Cutaneous Melanoma Patients for Immunotherapy and Targeted Therapy. Cancer Res 2021; 81:2545-2555. [PMID: 33574091 DOI: 10.1158/0008-5472.can-20-2000] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/22/2020] [Accepted: 02/09/2021] [Indexed: 11/16/2022]
Abstract
Malignant cutaneous melanoma is one of the most common cancers in young adults. During the last decade, targeted and immunotherapies have significantly increased the overall survival of patients with malignant cutaneous melanoma. Nevertheless, disease progression is common, and a lack of predictive biomarkers of patient response to therapy hinders individualized treatment strategies. To address this issue, we performed a longitudinal study using an unbiased proteomics approach to identify and quantify proteins in plasma both before and during treatment from 109 patients treated with either targeted or immunotherapy. Linear modeling and machine learning approaches identified 43 potential prognostic and predictive biomarkers. A reverse correlation between apolipoproteins and proteins related to inflammation was observed. In the immunotherapy group, patients with low pretreatment expression of apolipoproteins and high expression of inflammation markers had shorter progression-free survival. Similarly, increased expression of LDHB during treatment elicited a significant impact on response to immunotherapy. Overall, we identified potential common and treatment-specific biomarkers in malignant cutaneous melanoma, paving the way for clinical use of these biomarkers following validation on a larger cohort. SIGNIFICANCE: This study identifies a potential biomarker panel that could improve the selection of therapy for patients with cutaneous melanoma.
Collapse
Affiliation(s)
- Max J Karlsson
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | | | - Abdellah Tebani
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - David Kotol
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Veronica Höiom
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Linn Fagerberg
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Fredrik Edfors
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Mathias Uhlén
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | - Gianluca Maddalo
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
602
|
Kaneko A, Kanemaru H, Kajihara I, Mijiddorj T, Miyauchi H, Kuriyama H, Kimura T, Sawamura S, Makino K, Miyashita A, Aoi J, Makino T, Masuguchi S, Fukushima S, Ihn H. Liquid biopsy-based analysis by ddPCR and CAPP-Seq in melanoma patients. J Dermatol Sci 2021; 102:158-166. [PMID: 34049769 DOI: 10.1016/j.jdermsci.2021.04.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/22/2021] [Accepted: 04/25/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND The development of BRAF/MEK inhibitors in patients with metastatic melanoma harboring BRAF mutations has garnered attention for liquid biopsy to detect BRAF mutations in cell-free DNA (cfDNA) using droplet digital PCR (ddPCR) or next-generation sequencing methods. OBJECTIVE To investigate gene mutations in tumor DNA and cfDNA collected from 43 melanoma patients and evaluate their potential as biomarkers. METHODS ddPCR and CAncer Personalized Profiling by deep Sequencing (CAPP-Seq) techniques were performed to detect gene mutations in plasma cfDNA obtained from patients with metastatic melanoma. RESULTS Gene variants, including BRAF, NRAS, TP53, GNAS, and MET, were detectable in the plasma cfDNA, and the results were partially consistent with the results of those identified in the tissues. Among the variants examined, copy numbers of MET mutations were consistent with the disease status in two melanoma patients. CONCLUSION Liquid biopsy using CAPP-Seq and ddPCR has the potential to detect tumor presence and mutations, especially when tissue biopsies are unavailable. MET mutations in cfDNA may be a potential biomarker in patients with metastatic melanoma.
Collapse
Affiliation(s)
- Akira Kaneko
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hisashi Kanemaru
- 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
| | - Tselmeg Mijiddorj
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hitomi Miyauchi
- 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
| | - Toshihiro Kimura
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Soichiro Sawamura
- 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
| | - Azusa Miyashita
- 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
| | - Takamitsu Makino
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Shinichi Masuguchi
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoshi Fukushima
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hironobu Ihn
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
603
|
Pampena R, Michelini S, Lai M, Chester J, Pellacani G, Longo C. New systemic therapies for cutaneous melanoma: why, who and what. Ital J Dermatol Venerol 2021; 156:344-355. [PMID: 33913672 DOI: 10.23736/s2784-8671.21.06936-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Incidence of melanoma has been increasing in both sexes in the last decades. Advanced melanoma has always been one of the deadliest cancers worldwide due to his high metastatic capacity. In the last ten years, progresses in the knowledge of the molecular mechanisms involved in the melanoma development and progression, and in immune-response against melanoma, empowered the development of two new classes of systemic therapeutic agents: target-therapies and immunotherapies. Both classes consist of monoclonal antibodies inhibiting specific molecules. Target-therapies are selectively directed against cells harboring the BRAFV600-mutation, while immunotherapies target the two molecules involved in immune-checkpoint regulation, enhancing the immune response against the tumor: cytotoxic T-lymphocyte antigen-4 (CTLA-4) and programmed cell death-1 receptor (PD-1). Target- and immunotherapy demonstrated to improve both progression-free and overall survival in melanoma patients either in metastatic or in adjuvant settings. Several drugs have been approved in recent years as monotherapy or in combination, and many other drugs are currently under investigation in clinical trials. In the current review on new systemic therapies for cutaneous melanoma, we revised the molecular basis and the mechanisms of actions of both target- and immunotherapy (why). We discussed who are the best candidate to receive such therapies in both the adjuvant and metastatic setting (who) and which were the most important efficacy and safety data on these drugs (what).
Collapse
Affiliation(s)
- Riccardo Pampena
- Centro Oncologico ad Alta Tecnologia Diagnostica, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | | | - Michela Lai
- Centro Oncologico ad Alta Tecnologia Diagnostica, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, Reggio Emilia, Italy.,Department of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
| | - Johanna Chester
- Department of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
| | - Giovanni Pellacani
- Department of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
| | - Caterina Longo
- Centro Oncologico ad Alta Tecnologia Diagnostica, Azienda Unità Sanitaria Locale, IRCCS di Reggio Emilia, Reggio Emilia, Italy - .,Department of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
604
|
Wilkes JG, Patel A, McClure E, Pina Y, Zager JS. Developments in therapy for brain metastases in melanoma patients. Expert Opin Pharmacother 2021; 22:1443-1453. [PMID: 33688795 DOI: 10.1080/14656566.2021.1900117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Introduction: Cutaneous melanoma brain metastases (MBM) are a major cause of morbidity and mortality. While cytotoxic agents, interferon, or interleukin-2, have been used with some success in extracranial disease, limited efficacy is demonstrated in MBM. The rare patient with long-term survival presented with limited intracranial disease amenable to surgery or radiation therapy. However, the development of targeted therapy and immunotherapy over the last decade has significantly improved overall survival in this formerly devastating presentation of metastatic melanoma.Areas covered: This article reviews the mechanism of brain metastasis, challenges with treating the central nervous system, historical treatment of MBM, and outcomes in clinical trials with targeted therapy and immunotherapy.Expert opinion: The MBM patient population now, more than ever, requires a multidisciplinary approach with surgery, radiation therapy, and the use of newer systemic therapies such as immunotherapy agents and targeted therapy agents. MBM has traditionally been excluded from clinical trials for systemic therapy due to poor survival. However, recent data show overall survival rates have significantly improved, supporting the need for inclusion of MBM patients in systemic therapy clinical trials. Understanding the mechanisms of therapeutic activity in the brain, resistance mechanisms, and the appropriate multi-modality treatment approach requires further investigation. Nevertheless, these therapies continue to give some hope to patients with historically poor survival.
Collapse
Affiliation(s)
- Justin G Wilkes
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA.,University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Ayushi Patel
- University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Erin McClure
- University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Yolanda Pina
- Department of Neuro-Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Jonathan S Zager
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA.,University of South Florida Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
605
|
Cheng LY, Haydu LE, Song P, Nie J, Tetzlaff MT, Kwong LN, Gershenwald JE, Davies MA, Zhang DY. High sensitivity sanger sequencing detection of BRAF mutations in metastatic melanoma FFPE tissue specimens. Sci Rep 2021; 11:9043. [PMID: 33907234 PMCID: PMC8079675 DOI: 10.1038/s41598-021-88391-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 04/08/2021] [Indexed: 01/17/2023] Open
Abstract
Mutations in the BRAF gene at or near the p. V600 locus are informative for therapy selection, but current methods for analyzing FFPE tissue DNA generally have a limit of detection of 5% variant allele frequency (VAF), or are limited to the single variant (V600E). These can result in false negatives for samples with low VAFs due to low tumor content or subclonal heterogeneity, or harbor non-V600 mutations. Here, we show that Sanger sequencing using the NuProbe VarTrace BRAF assay, based on the Blocker Displacement Amplification (BDA) technology, is capable of detecting BRAF V600 mutations down to 0.20% VAF from FFPE lymph node tissue samples. Comparison experiments on adjacent tissue sections using BDA Sanger, immunohistochemistry (IHC), digital droplet PCR (ddPCR), and NGS showed 100% concordance among all 4 methods for samples with BRAF mutations at ≥ 1% VAF, though ddPCR did not distinguish the V600K mutation from the V600E mutation. BDA Sanger, ddPCR, and NGS (with orthogonal confirmation) were also pairwise concordant for lower VAF mutations down to 0.26% VAF, but IHC produced a false negative. Thus, we have shown that Sanger sequencing can be effective for rapid detection and quantitation of multiple low VAF BRAF mutations from FFPE samples. BDA Sanger method also enabled detection and quantitation of less frequent, potentially actionable non-V600 mutations as demonstrated by synthetic samples.
Collapse
Affiliation(s)
- Lauren Y Cheng
- Department of Bioengineering, Rice University, 65000 Main St, Houston, TX, 77030, USA
| | - Lauren E Haydu
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Ping Song
- Department of Bioengineering, Rice University, 65000 Main St, Houston, TX, 77030, USA
| | - Jianyi Nie
- Department of Bioengineering, Rice University, 65000 Main St, Houston, TX, 77030, USA
| | - Michael T Tetzlaff
- Department of Translational Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Lawrence N Kwong
- Department of Translational Molecular Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Michael A Davies
- Department of Melanoma Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - David Yu Zhang
- Department of Bioengineering, Rice University, 65000 Main St, Houston, TX, 77030, USA.
- Systems, Synthetic, and Physical Biology, Rice University, Houston, TX, USA.
| |
Collapse
|
606
|
Real-World Survival in Patients with Metastatic Melanoma after Discontinuation of Anti-PD-1 Immunotherapy for Objective Response or Adverse Effects: A Retrospective Study. JOURNAL OF ONCOLOGY 2021; 2021:5524685. [PMID: 33995528 PMCID: PMC8096591 DOI: 10.1155/2021/5524685] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/25/2021] [Accepted: 04/09/2021] [Indexed: 01/05/2023]
Abstract
Objective Anti-PD-1 has dramatically improved the survival of patients with advanced melanoma. However, there is a lack of data on maintenance of the response after treatment discontinuation. We aimed to evaluate the progression-free survival (PFS) of patients with metastatic melanoma after anti-PD-1 interruption for objective response (OR) or limiting toxicity during clinical trials. Methods All patients with advanced melanoma who stopped single-agent anti-PD-1 antibodies for objective response or toxicity were included between April 2014 and January 2019 in our institution (data cut-off, September 10th, 2019). Clinical and biological factors associated with relapse were studied. Results The median follow-up after introduction of treatment was 36.5 months [4.6–62.4], and the median follow-up after discontinuation of treatment was 15.7 months (2.5–45.1). Out of 65 patients, 28 patients stopped immunotherapy for limiting adverse effects (AEs) (43.1%), 25 for complete response (CR) (38.4%), and 12 for partial response (PR) or long-term stable disease (SD) (18.5%). Twelve patients relapsed (18.5%) after a median time of 9 months [1.9–40.9 months]. Seven relapsed after discontinuation for AEs, 3 after discontinuation for CR, and 2 after discontinuation for PR/SD. The median PFS after therapy discontinuation was not reached. No statistical association was found between recurrence and age, sex, increased LDH, BRAF status, presence of brain metastases, previous treatments, radiotherapy, or time on anti-PD-1 treatment. Conclusion This cohort shows a global recurrence rate of 18.5% and confirms a long-lasting response after anti-PD-1 cessation regardless of the cause of discontinuation.
Collapse
|
607
|
Argenziano G, Brancaccio G, Moscarella E, Dika E, Fargnoli MC, Ferrara G, Longo C, Pellacani G, Peris K, Pimpinelli N, Quaglino P, Rongioletti F, Simonacci M, Zalaudek I, Calzavara Pinton P. Management of cutaneous melanoma: comparison of the leading international guidelines updated to the 8th American Joint Committee on Cancer staging system and workup proposal by the Italian Society of Dermatology. GIORN ITAL DERMAT V 2021; 155:126-145. [PMID: 32394673 DOI: 10.23736/s0392-0488.19.06383-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Giuseppe Argenziano
- Unit of Dermatology, Luigi Vanvitelli University of Campania, Naples, Italy -
| | | | - Elvira Moscarella
- Unit of Dermatology, Luigi Vanvitelli University of Campania, Naples, Italy
| | - Emi Dika
- Unit of Dermatology (DIMES), University of Bologna, Bologna, Italy
| | - Maria C Fargnoli
- Department of Dermatology, University of L'Aquila, L'Aquila, Italy
| | - Gerardo Ferrara
- Unit of Anatomic Pathology, Hospital of Macerata, Area Vasta 3 ASUR Marche, Macerata, Italy
| | - Caterina Longo
- Department of Dermatology, University of Modena and Reggio Emilia, Modena, Italy.,Azienda Unità Sanitaria Locale - IRCCS di Reggio Emilia, Centro Oncologico ad Alta Tecnologia Diagnostica-Dermatologia, Reggio Emilia, Italy
| | - Giovanni Pellacani
- Department of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
| | - Ketty Peris
- Institute of Dermatology, Sacred Heart Catholic University, Rome, Italy.,A. Gemelli University Polyclinic, IRCCS and Foundation, Rome, Italy
| | - Nicola Pimpinelli
- Unit of Dermatology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Pietro Quaglino
- Dermatologic Clinic, Department of Medical Sciences, University of Turin Medical School, Turin, Italy
| | - Franco Rongioletti
- Unit of Dermatology, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Marco Simonacci
- Unit of Dermatology, Hospital of Macerata, Area Vasta 3 ASUR Marche, Macerata, Italy
| | - Iris Zalaudek
- Department of Dermatology, University Hospital of Trieste, Trieste, Italy
| | | |
Collapse
|
608
|
Gogebakan KC, Mukherjee K, Berry EG, Sonmez K, Leachman SA, Etzioni R. Impact of novel systemic therapies on the first-year costs of care for melanoma among Medicare beneficiaries. Cancer 2021; 127:2926-2933. [PMID: 33905529 DOI: 10.1002/cncr.33515] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 02/14/2021] [Accepted: 02/15/2021] [Indexed: 01/22/2023]
Abstract
BACKGROUND Since 2011, the therapeutic landscape of melanoma has changed dramatically because of the adoption of immune checkpoint inhibitor and targeted therapies. The authors sought to quantify the effects of these changes on short-term treatment costs by comparing the first-year cancer-attributable costs in novel (2011-2015) and historical (2004-2010) treatment eras. METHODS The authors estimated the first-year cancer-attributable and out-of-pocket (OOP) costs by cancer stage at diagnosis by using a case-control approach. Patients aged ≥67 years with melanoma results were used to calculate the total direct costs of treatment during the first year after the diagnosis of melanoma in the US Medicare population older than 65 years. Costs were reported in 2018 dollars. RESULTS Costs increased with the stage at diagnosis. Average first-year cancer-attributable costs per patient for stage IV patients increased significantly by 61.7% from $45,952 to $74,297 after the adoption of novel treatments. Per-patient OOP responsibility decreased by almost 30.8% across all stages of cancer but increased by 16.5% for stage IV patients from 2004 ($7646) to 2015 ($8911). The total direct cost of treatment for persons with melanoma older than 65 years increased by $16.03 million (4.93%) from $324.68 million in 2010 to $340.71 million in 2015. The largest increase in yearly total cost, $23.64 million (56.53%), was observed among stage IV patients. CONCLUSIONS The direct cost of melanoma increased significantly in the Medicare population, particularly for advanced-stage disease. Prevention and early detection initiatives may reduce the economic burden of melanoma.
Collapse
Affiliation(s)
- Kemal Caglar Gogebakan
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Kumar Mukherjee
- School of Pharmacy, Philadelphia College of Osteopathic Medicine, Suwanee, Georgia
| | - Elizabeth G Berry
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon
| | - Kemal Sonmez
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Sancy A Leachman
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon
| | - Ruth Etzioni
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Statistics, University of Washington, Seattle, Washington
| |
Collapse
|
609
|
Yang A, Li M, Fang M. The Research Progress of Direct KRAS G12C Mutation Inhibitors. Pathol Oncol Res 2021; 27:631095. [PMID: 34257597 PMCID: PMC8262225 DOI: 10.3389/pore.2021.631095] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/01/2021] [Indexed: 11/26/2022]
Abstract
Abstract: KRAS mutations have long been considered undruggable. However, a series of direct KRAS mutation inhibitors have been developed since the switch II pocket was discovered recently. This review will summarize progress in the development of direct KRAS G12C mutation inhibitors, current relevant drugs under study and challenges that need to be considered in future research.
Collapse
Affiliation(s)
- Ai Yang
- Department of Oncology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Min Li
- Department of Oncology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Mingzhi Fang
- Department of Oncology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
610
|
Raza M, Kumar N, Nair U, Luthra G, Bhattacharyya U, Jayasundar S, Jayasundar R, Sehrawat S. Current updates on precision therapy for breast cancer associated brain metastasis: Emphasis on combination therapy. Mol Cell Biochem 2021; 476:3271-3284. [PMID: 33886058 DOI: 10.1007/s11010-021-04149-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 04/01/2021] [Indexed: 12/12/2022]
Abstract
Cancer therapies have undergone a tremendous progress over the past decade. Precision medicine provides a more tailored approach, making the combination of existing therapies more precise. Different types of cancers are characterized by unique biomarkers that are targeted using various genomic approaches by clinicians and companies worldwide to achieve efficient treatment with minimal side effects. Precision medicine has two broad approaches namely stratified and personalized medicine. The driver mutations could vary within a subtype while the same driver mutations could be found across different subtypes. Precision medicine has recently gained a lot of importance for breast cancer therapy. Various kinds of mutations like hotspot mutations, gene alterations, gene amplification mutations are targeted to design a more specific therapy. Apart from these known gene mutations there are various unknown mutations. Thus, tumor heterogeneity can pose a challenge to precision medicine. For breast cancer, one of the most successful models developed in case of precision medicine is the anti-HER2 therapies as HER2 was considered to have the worst prognosis being highly malignant. But now due to the advent of HER2 receptor targeted therapies, it has a good prognosis. Moreover, precision medicine helps in identifying if the drug molecules being used for the treatment of one kind of cancer can be beneficial in the treatment of another kind of cancer as well, considering the signaling pathways and machinery is similar in most of the cancers. This reduces the time for new drug development and is economically more feasible. Precision medicine will prove to be very advantageous in case of brain metastasis.
Collapse
Affiliation(s)
- Masoom Raza
- Precision NeuroOncology & NeuroVascular Disease Modeling Group, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Delhi NCR, India
| | - Naveen Kumar
- Precision NeuroOncology & NeuroVascular Disease Modeling Group, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Delhi NCR, India
| | - Uttara Nair
- Department of Women's and Reproductive Health, Oxford Fertility, Oxford Business Park North, University of Oxford, Oxford, OX4 2HW, UK
| | - Gehna Luthra
- Precision NeuroOncology & NeuroVascular Disease Modeling Group, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Delhi NCR, India
| | - Ushosi Bhattacharyya
- Precision NeuroOncology & NeuroVascular Disease Modeling Group, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Delhi NCR, India
| | - Smruthi Jayasundar
- Precision NeuroOncology & NeuroVascular Disease Modeling Group, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Delhi NCR, India
| | - Rama Jayasundar
- Department of Nuclear Magnetic Resonance & MRI, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Seema Sehrawat
- Precision NeuroOncology & NeuroVascular Disease Modeling Group, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Delhi NCR, India.
| |
Collapse
|
611
|
Yu SY, Mckavanagh D, McPherson I, Walpole E, Atkinson V, Hollingworth S. Survival of advanced melanoma patients treated with immunotherapy and targeted therapy: A real-world study. Pharmacoepidemiol Drug Saf 2021; 30:1371-1379. [PMID: 33840147 DOI: 10.1002/pds.5248] [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: 10/28/2020] [Revised: 04/07/2021] [Accepted: 04/09/2021] [Indexed: 11/09/2022]
Abstract
INTRODUCTION We aimed to examine the survival outcomes plus patient and treatment characteristics of advanced melanoma patients treated with first-line immunotherapy (IT), targeted therapy (TT), and chemotherapy (CTH) and compare findings with information from pivotal trials for each therapy. MATERIALS AND METHODS We retrospectively reviewed the use of systematic IT, TT and CTH therapies in melanoma patients in four Queensland public hospitals. We estimated median duration of overall survival (OS) and survival rates (6 months, 1, and 2 years) using Kaplan-Meier methods. We compared our findings to those of clinical trials. RESULTS Five hundred three patients who met the inclusion criteria were divided into three groups based on the first-line treatment: IT 232; TT 157; and CTH 114. OS was 18 months with IT (95% CI 13, 22); 12 months with TT (95% CI 8, 15); and 5 months with CTH (95% CI 5, 6). The demographic characteristics, treatment protocols, and durations for IT and TT were generally consistent with trials but fewer patients in our study had subsequent therapy than in the trials. The OS in our study was slightly lower than the OS reported in trials. CONCLUSION The OS of novel cancer therapy in the real world was lower than seen in trials but is expected given these are patients who have a poorer prognosis. A future study could investigate the impact of prognostic factors on survival in the longer term. This study provides evidence that we can use routinely collected real-world data to evaluate the effectiveness of checkpoint and kinase inhibitors in patients with advanced melanoma.
Collapse
Affiliation(s)
- Su-Yeon Yu
- National Evidence-based Healthcare Collaborating Agency, Seoul, South Korea
| | - Dan Mckavanagh
- Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Ian McPherson
- Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Euan Walpole
- Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | | | | |
Collapse
|
612
|
Hoang VT, Matossian MD, La J, Hoang K, Ucar DA, Elliott S, Burks HE, Wright TD, Patel S, Bhatt A, Phamduy T, Chrisey D, Buechlein A, Rusch DB, Nephew KP, Anbalagan M, Rowan B, Cavanaugh JE, Flaherty PT, Miele L, Collins-Burow BM, Burow ME. Dual inhibition of MEK1/2 and MEK5 suppresses the EMT/migration axis in triple-negative breast cancer through FRA-1 regulation. J Cell Biochem 2021; 122:835-850. [PMID: 33876843 DOI: 10.1002/jcb.29916] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/22/2021] [Accepted: 02/25/2021] [Indexed: 01/03/2023]
Abstract
Triple-negative breast cancer (TNBC) presents a clinical challenge due to the aggressive nature of the disease and a lack of targeted therapies. Constitutive activation of the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway has been linked to chemoresistance and metastatic progression through distinct mechanisms, including activation of epithelial-to-mesenchymal transition (EMT) when cells adopt a motile and invasive phenotype through loss of epithelial markers (CDH1), and acquisition of mesenchymal markers (VIM, CDH2). Although MAPK/ERK1/2 kinase inhibitors (MEKi) are useful antitumor agents in a clinical setting, including the Food and Drug Administration (FDA)-approved MEK1,2 dual inhibitors cobimetinib and trametinib, there are limitations to their clinical utility, primarily adaptation of the BRAF pathway and ocular toxicities. The MEK5 (HGNC: MAP2K5) pathway has important roles in metastatic progression of various cancer types, including those of the prostate, colon, bone and breast, and elevated levels of ERK5 expression in breast carcinomas are linked to a worse prognoses in TNBC patients. The purpose of this study is to explore MEK5 regulation of the EMT axis and to evaluate a novel pan-MEK inhibitor on clinically aggressive TNBC cells. Our results show a distinction between the MEK1/2 and MEK5 cascades in maintenance of the mesenchymal phenotype, suggesting that the MEK5 pathway may be necessary and sufficient in EMT regulation while MEK1/2 signaling further sustains the mesenchymal state of TNBC cells. Furthermore, additive effects on MET induction are evident through the inhibition of both MEK1/2 and MEK5. Taken together, these data demonstrate the need for a better understanding of the individual roles of MEK1/2 and MEK5 signaling in breast cancer and provide a rationale for the combined targeting of these pathways to circumvent compensatory signaling and subsequent therapeutic resistance.
Collapse
Affiliation(s)
- Van T Hoang
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Margarite D Matossian
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Jacqueline La
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Kristine Hoang
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Deniz A Ucar
- Department of Genetics and Stanley S. Scott Cancer Center, LSUHSC, New Orleans, Louisiana, USA
| | - Steven Elliott
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Hope E Burks
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Thomas D Wright
- Department of Pharmacology, Duquesne University, School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Saloni Patel
- Department of Pharmacology, Duquesne University, School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Akshita Bhatt
- Department of Pharmacology, Duquesne University, School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Theresa Phamduy
- Department of Physics, Tulane University, New Orleans, Louisiana, USA
| | - Douglas Chrisey
- Department of Physics, Tulane University, New Orleans, Louisiana, USA
| | - Aaron Buechlein
- Medical Sciences Program, Indiana University School of Medicine-Bloomington, Bloomington, Indiana, USA
| | - Douglas B Rusch
- Center for Genomics and Bioinformatics, Indiana University, Bloomington, Indiana, USA
| | - Kenneth P Nephew
- Medical Sciences Program, Indiana University School of Medicine-Bloomington, Bloomington, Indiana, USA
| | - Murali Anbalagan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Brian Rowan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Jane E Cavanaugh
- Department of Pharmacology, Duquesne University, School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Patrick T Flaherty
- Department of Medicinal Chemistry, Duquesne University, School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Lucio Miele
- Department of Genetics and Stanley S. Scott Cancer Center, LSUHSC, New Orleans, Louisiana, USA
| | - Bridgette M Collins-Burow
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, Louisiana, USA.,Tulane Cancer Center, New Orleans, Louisiana, USA
| | - Matthew E Burow
- Department of Medicine, Section of Hematology and Oncology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
613
|
Wenzel C, Herold S, Wermke M, E. Aust D, B. Baretton G. Routine Molecular Pathology Diagnostics in Precision Oncology. DEUTSCHES ARZTEBLATT INTERNATIONAL 2021; 118:arztebl.m2021.0025. [PMID: 33536117 PMCID: PMC8287073 DOI: 10.3238/arztebl.m2021.0025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 12/01/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND Technical advances in the field of molecular genetics permit precise genomic characterization of malignant tumors. This has not only improved our understanding of tumor biology but also paved the way for molecularly stratified treatment strategies in routine clinical practice. METHODS A selective search of PubMed to identify literature on molecular pathology methods, their indications, the challenges associated with molecular findings, and future developments. RESULTS Tumors can be characterized with the aid of immunohistochemistry, in-situ hybridization, and sequencing of DNA or RNA. The benefits of molecularly stratified tumor treatment have been demonstrated by randomized clinical trials on numerous tumor entities, e.g., non-small-cell lung cancer, colorectal cancer, and breast cancer. Therefore, initiation of specific treatment for these entities should be preceded by molecular pathology biomarker analyses, generally carried out on tumor tissue. Randomized controlled trials and non-controlled studies show that enhanced progression-free survival ensues if the pharmacological treatment is oriented on the findings of molecular pathology diagnostics. In next-generation sequencing, numerous relevant gene sequences or even whole genes can be sequenced in parallel, dispensing with complex staged diagnostics and reducing the use of biomaterials. These new methods also complement the currently relevant predictive biomarkers by permitting the investigation of genetic alterations presently of interest in the context of clinical studies. Prior to widespread routine clinical application, however, sequencing of large gene panels or whole genomes or exomes need to be even more stringently validated. CONCLUSION Quality-assured molecular pathology assays are universally available for the determination of currently relevant predictive biomarkers. However, the integration of extensive genomic analyses into routine molecular pathology diagnostics represents a future challenge in precision oncology.
Collapse
Affiliation(s)
- Carina Wenzel
- Institute of Pathology, University Hospital Carl Gustav Carus Dresden, TU Dresden, Dresden
| | - Sylvia Herold
- Institute of Pathology, University Hospital Carl Gustav Carus Dresden, TU Dresden, Dresden
| | - Martin Wermke
- Medical Department I, University Hospital Carl Gustav Carus Dresden, TU Dresden, Dresden: Dr. med. Martin Wermke
| | - Daniela E. Aust
- Institute of Pathology, University Hospital Carl Gustav Carus Dresden, TU Dresden, Dresden
| | - Gustavo B. Baretton
- Institute of Pathology, University Hospital Carl Gustav Carus Dresden, TU Dresden, Dresden
| |
Collapse
|
614
|
Boulva K, Apte S, Yu A, Tran A, Shorr R, Song X, Ong M, Nessim C. Contemporary Neoadjuvant Therapies for High-Risk Melanoma: A Systematic Review. Cancers (Basel) 2021; 13:1905. [PMID: 33920967 PMCID: PMC8071293 DOI: 10.3390/cancers13081905] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/10/2021] [Accepted: 04/13/2021] [Indexed: 11/16/2022] Open
Abstract
Despite advances in adjuvant immuno- and targeted therapies, the risk of relapse for stage III melanoma remains high. With 43 active entries on clinicaltrials.gov (8 July 2020), there is a surge of interest in the role of contemporary therapies in the neoadjuvant setting. We conducted a systematic review of trials performed in the last decade evaluating neoadjuvant targeted, immuno- or intralesional therapy for resectable stage III or IV melanoma. Database searches of Medline, Embase, and the Cochrane Central Register of Controlled Trials were conducted from inception to 13 February 2020. Two reviewers assessed titles, abstracts, and full texts. Trials investigating contemporary neoadjuvant therapies in high-risk melanoma were included. Eight phase II trials (4 randomized and 4 single-arm) involving 450 patients reported on neoadjuvant anti-BRAF/MEK targeted therapy (3), anti-PD-1/CTLA-4 immunotherapy (3), and intralesional therapy (2). The safest and most efficacious regimens were dabrafenib/trametinib and combination ipilimumab (1 mg/kg) + nivolumab (3 mg/kg). Pathologic complete response (pCR) and adverse events were comparable. Ipilimumab + nivolumab exhibited longer RFS. Contemporary neoadjuvant therapies are not only safe, but also demonstrate remarkable pCR and RFS-outcomes which are regarded as meaningful surrogates for long-term survival. Studies defining predictors of pCR, its correlation with oncologic outcomes, and phase III trials comparing neoadjuvant therapy to standard of care will be crucial.
Collapse
Affiliation(s)
- Kerianne Boulva
- Division of General Surgery, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (K.B.); (S.A.); (A.T.)
| | - Sameer Apte
- Division of General Surgery, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (K.B.); (S.A.); (A.T.)
- The Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (R.S.); (X.S.); (M.O.)
| | - Ashley Yu
- Department of Family Medicine, McMaster University, Hamilton, ON L8P 1H6, Canada;
| | - Alexandre Tran
- Division of General Surgery, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (K.B.); (S.A.); (A.T.)
- The Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (R.S.); (X.S.); (M.O.)
| | - Risa Shorr
- The Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (R.S.); (X.S.); (M.O.)
| | - Xinni Song
- The Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (R.S.); (X.S.); (M.O.)
- Division of Medical Oncology, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| | - Michael Ong
- The Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (R.S.); (X.S.); (M.O.)
- Division of Medical Oncology, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
| | - Carolyn Nessim
- Division of General Surgery, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada; (K.B.); (S.A.); (A.T.)
- The Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; (R.S.); (X.S.); (M.O.)
| |
Collapse
|
615
|
Li Y, Umbach DM, Krahn JM, Shats I, Li X, Li L. Predicting tumor response to drugs based on gene-expression biomarkers of sensitivity learned from cancer cell lines. BMC Genomics 2021; 22:272. [PMID: 33858332 PMCID: PMC8048084 DOI: 10.1186/s12864-021-07581-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 04/04/2021] [Indexed: 02/07/2023] Open
Abstract
Background Human cancer cell line profiling and drug sensitivity studies provide valuable information about the therapeutic potential of drugs and their possible mechanisms of action. The goal of those studies is to translate the findings from in vitro studies of cancer cell lines into in vivo therapeutic relevance and, eventually, patients’ care. Tremendous progress has been made. Results In this work, we built predictive models for 453 drugs using data on gene expression and drug sensitivity (IC50) from cancer cell lines. We identified many known drug-gene interactions and uncovered several potentially novel drug-gene associations. Importantly, we further applied these predictive models to ~ 17,000 bulk RNA-seq samples from The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) database to predict drug sensitivity for both normal and tumor tissues. We created a web site for users to visualize and download our predicted data (https://manticore.niehs.nih.gov/cancerRxTissue). Using trametinib as an example, we showed that our approach can faithfully recapitulate the known tumor specificity of the drug. Conclusions We demonstrated that our approach can predict drugs that 1) are tumor-type specific; 2) elicit higher sensitivity from tumor compared to corresponding normal tissue; 3) elicit differential sensitivity across breast cancer subtypes. If validated, our prediction could have relevance for preclinical drug testing and in phase I clinical design. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07581-7.
Collapse
Affiliation(s)
- Yuanyuan Li
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, 111 T.W. Alexander Dr., Research Triangle Park, MD A3-03, Durham, NC, 27709, USA
| | - David M Umbach
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, 111 T.W. Alexander Dr., Research Triangle Park, MD A3-03, Durham, NC, 27709, USA
| | - Juno M Krahn
- Genome Integrity & Structural Biology Laboratory, Research Triangle Park, Durham, NC, 27709, USA
| | - Igor Shats
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, 27709, USA
| | - Xiaoling Li
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, 27709, USA
| | - Leping Li
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, 111 T.W. Alexander Dr., Research Triangle Park, MD A3-03, Durham, NC, 27709, USA.
| |
Collapse
|
616
|
Zeman J, Olivová L, Hrudka J, Hajer J, Rychlík I. Obstructive Jaundice Secondary to Pancreatic Head Metastasis of Malignant Amelanotic Melanoma as the First Clinical Manifestation. Prague Med Rep 2021; 122:45-51. [PMID: 33646942 DOI: 10.14712/23362936.2021.6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Malignant melanoma is commonly known for its high probability of metastasizing to distant organs. Metastases to gastrointestinal tract are well documented, but resulting jaundice is only scarcely seen. We present a case of histologically verified metastasis of amelanotic melanoma to the head of pancreas infiltrating the common bile duct and consequently causing obstructive jaundice which constituted its first clinical manifestation. Multidisciplinary approach is essential in patients with malignant melanoma since early detection of the melanoma or its metastases may improve patients' clinical outcome, especially owing to the use of targeted biological treatment without any delay.
Collapse
Affiliation(s)
- Jan Zeman
- Department of Internal Medicine, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czech Republic.
| | - Lucie Olivová
- Department of Dermatology, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czech Republic
| | - Jan Hrudka
- Department of Pathology, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czech Republic
| | - Jan Hajer
- Department of Gastroenterology, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czech Republic
| | - Ivan Rychlík
- Department of Internal Medicine, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czech Republic
| |
Collapse
|
617
|
Huestis MP, Dela Cruz D, DiPasquale AG, Durk MR, Eigenbrot C, Gibbons P, Gobbi A, Hunsaker TL, La H, Leung DH, Liu W, Malek S, Merchant M, Moffat JG, Muli CS, Orr CJ, Parr BT, Shanahan F, Sneeringer CJ, Wang W, Yen I, Yin J, Siu M, Rudolph J. Targeting KRAS Mutant Cancers via Combination Treatment: Discovery of a 5-Fluoro-4-(3 H)-quinazolinone Aryl Urea pan-RAF Kinase Inhibitor. J Med Chem 2021; 64:3940-3955. [PMID: 33780623 DOI: 10.1021/acs.jmedchem.0c02085] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Optimization of a series of aryl urea RAF inhibitors led to the identification of type II pan-RAF inhibitor GNE-0749 (7), which features a fluoroquinazolinone hinge-binding motif. By minimizing reliance on common polar hinge contacts, this hinge binder allows for a greater contribution of RAF-specific residue interactions, resulting in exquisite kinase selectivity. Strategic substitution of fluorine at the C5 position efficiently masked the adjacent polar NH functionality and increased solubility by impeding a solid-state conformation associated with stronger crystal packing of the molecule. The resulting improvements in permeability and solubility enabled oral dosing of 7. In vivo evaluation of 7 in combination with the MEK inhibitor cobimetinib demonstrated synergistic pathway inhibition and significant tumor growth inhibition in a KRAS mutant xenograft mouse model.
Collapse
Affiliation(s)
- Malcolm P Huestis
- Discovery Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Darlene Dela Cruz
- Translational Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Antonio G DiPasquale
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Matthew R Durk
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Charles Eigenbrot
- Structural Biology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Paul Gibbons
- Discovery Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Alberto Gobbi
- Discovery Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Thomas L Hunsaker
- Translational Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Hank La
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Dennis H Leung
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Wendy Liu
- Discovery Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Shiva Malek
- Molecular Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Mark Merchant
- Translational Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - John G Moffat
- Biochemical and Cellular Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Christine S Muli
- Small Molecule Pharmaceutical Sciences, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Christine J Orr
- Translational Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Brendan T Parr
- Discovery Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Frances Shanahan
- Molecular Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Christopher J Sneeringer
- Biochemical and Cellular Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Weiru Wang
- Structural Biology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Ivana Yen
- Molecular Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jianping Yin
- Structural Biology, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Michael Siu
- Discovery Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Joachim Rudolph
- Discovery Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
618
|
Kummar S, Berlin J, Mascarenhas L, van Tilburg CM, Geoerger B, Lassen UN, Schilder RJ, Turpin B, Nanda S, Keating K, Childs BH, Chirila C, Laetsch TW, Hyman DM, Drilon A, Hong DS. Quality of Life in Adult and Pediatric Patients with Tropomyosin Receptor Kinase Fusion Cancer Receiving Larotrectinib. Curr Probl Cancer 2021; 45:100734. [PMID: 33865615 DOI: 10.1016/j.currproblcancer.2021.100734] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/18/2021] [Accepted: 03/02/2021] [Indexed: 01/08/2023]
Abstract
Neurotrophic tyrosine receptor kinase (NTRK) gene fusions lead to chimeric tropomyosin receptor kinase (TRK) fusion proteins, which act as primary oncogenic drivers in diverse tumor types in adults and children. Larotrectinib, a highly selective and central nervous system-active TRK inhibitor, has shown high objective response rates, durable disease control, and a favorable safety profile in patients with TRK fusion cancer. The impact of larotrectinib on health-related quality of life (HRQoL) was evaluated in adult and pediatric patients in two phase I/II clinical trials (NAVIGATE; NCT02576431 and SCOUT; NCT02637687). Patients completed HRQoL questionnaires (EORTC QLQ-C30, EQ-5D-5L, and PedsQL) at baseline and at planned treatment cycle visits. Changes in questionnaire scores were evaluated over time, and by tumor type and treatment response. Questionnaires from 40 adult and 17 pediatric (2-19 years of age) patients receiving larotrectinib were completed at baseline and at least one post-baseline timepoint. Meaningful within-patient HRQoL improvements occurred at one or more timepoints in 60% of adults and 76% of pediatric patients. Sustained improvements in EORTC QLQ-C30 and PedsQL scores were rapid, occurring within 2 months of treatment initiation in 68% and 71% of patients, respectively. Improvements were observed regardless of tumor type and appeared to correlate with clinical efficacy. The rapid within-patient HRQoL improvements in adult and pediatric patients with TRK fusion cancer are consistent with the clinical profile of larotrectinib. Our results provide valuable information for use of this agent in this patient population. A plain language summary of this article is available in the supplementary appendix.
Collapse
Affiliation(s)
- Shivaani Kummar
- Stanford Cancer Institute, Stanford University, Palo Alto, CA.
| | | | - Leo Mascarenhas
- Cancer and Blood Disease Institute, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Cornelis M van Tilburg
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg University Hospital and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Birgit Geoerger
- Gustave Roussy Cancer Center, Department of Pediatric and Adolescent Oncology, Université Paris-Saclay, Villejuif, France
| | - Ulrik N Lassen
- Department of Oncology, Rigshospitalet, Copenhagen, Denmark
| | - Russell J Schilder
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Brian Turpin
- Division of Pediatric Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Shivani Nanda
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, NJ
| | - Karen Keating
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, NJ
| | | | | | - Theodore W Laetsch
- Department of Pediatrics and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center/Children's Health, Dallas, TX
| | - David M Hyman
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - Alexander Drilon
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - David S Hong
- University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
619
|
Aldea M, Andre F, Marabelle A, Dogan S, Barlesi F, Soria JC. Overcoming Resistance to Tumor-Targeted and Immune-Targeted Therapies. Cancer Discov 2021; 11:874-899. [PMID: 33811122 DOI: 10.1158/2159-8290.cd-20-1638] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/13/2021] [Accepted: 02/01/2021] [Indexed: 11/16/2022]
Abstract
Resistance to anticancer therapies includes primary resistance, usually related to lack of target dependency or presence of additional targets, and secondary resistance, mostly driven by adaptation of the cancer cell to the selection pressure of treatment. Resistance to targeted therapy is frequently acquired, driven by on-target, bypass alterations, or cellular plasticity. Resistance to immunotherapy is often primary, orchestrated by sophisticated tumor-host-microenvironment interactions, but could also occur after initial efficacy, mostly when only partial responses are obtained. Here, we provide an overview of resistance to tumor and immune-targeted therapies and discuss challenges of overcoming resistance, and current and future directions of development. SIGNIFICANCE: A better and earlier identification of cancer-resistance mechanisms could avoid the use of ineffective drugs in patients not responding to therapy and provide the rationale for the administration of personalized drug associations. A clear description of the molecular interplayers is a prerequisite to the development of novel and dedicated anticancer drugs. Finally, the implementation of such cancer molecular and immunologic explorations in prospective clinical trials could de-risk the demonstration of more effective anticancer strategies in randomized registration trials, and bring us closer to the promise of cure.
Collapse
Affiliation(s)
- Mihaela Aldea
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Fabrice Andre
- Department of Medical Oncology, Gustave Roussy, Villejuif, France.,INSERM U981, PRISM Institute, Gustave Roussy, Villejuif, France.,Paris Saclay University, Saint-Aubin, France
| | - Aurelien Marabelle
- INSERM U981, PRISM Institute, Gustave Roussy, Villejuif, France.,Drug Development Department, Gustave Roussy, Villejuif, France
| | - Semih Dogan
- INSERM U981, PRISM Institute, Gustave Roussy, Villejuif, France
| | - Fabrice Barlesi
- Department of Medical Oncology, Gustave Roussy, Villejuif, France.,Aix Marseille University, CNRS, INSERM, CRCM, Marseille, France
| | - Jean-Charles Soria
- Paris Saclay University, Saint-Aubin, France. .,Drug Development Department, Gustave Roussy, Villejuif, France
| |
Collapse
|
620
|
Liu X, Li JJ, Ding Y, Li DD, Wen XZ, Weng DS, Wang JH, Jiang H, Zhang XS. Safety and Tolerability of BRAF Inhibitor and BRAF Inhibitor-Based Combination Therapy in Chinese Patients With Advanced Melanoma: A Real World Study. Front Oncol 2021; 11:582676. [PMID: 33868987 PMCID: PMC8047100 DOI: 10.3389/fonc.2021.582676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 02/12/2021] [Indexed: 11/13/2022] Open
Abstract
The toxicity spectrum between Chinese and Caucasian patients with melanoma who were treated with BRAF inhibitors (BRAFi) may differ. The purpose of the present study was to assess the safety and tolerability of BRAFi and BRAFi-based combination therapies [MEK inhibitors (MEKi) or anti-programmed death-1 (PD-1) antibody] in Chinese patients with BRAF V600E/K mutation-positive metastatic melanoma. We also investigated whether treatment-related adverse events (AEs) correlated with the prognosis. This retrospective study collected data from 43 patients with BRAF V600E/K mutation-positive metastatic melanoma from a single Chinese cancer center. Of the 43 patients, 12 patients received BRAFi monotherapy, 12 patients received BRAFi+MEKi, and 19 patients received BRAFi combined with the anti-PD-1 antibody. The median follow-up time was 19 months. In the BRAFi group, the most common AEs were rashes, palmoplantar erythrodysesthesia, and arthralgia. Four out of 12 (30%) patients experienced grade 3–4 treatment-related AEs. All grades of AEs in the BRAFi+MEKi group were similar to the BRAFi group, except for higher pyrexia (58.3%) and fewer cutaneous AEs. Three out of 12 (25%) patients experienced grade 3–4 AEs, especially pyrexia (16.7%). In the BRAFi+anti-PD-1 antibody group, AEs were similar to the BRAFi group, except for an increased aminotransferase level (36.8%), increased bilirubin (31.6%), and hypothyroidism (15.8%). Eleven out of 19 (57.9%) patients experienced grade 3–4 AEs and four out of 19 (21%) patients discontinued the therapy due to AEs. Treatment-related hepatotoxicity (trHE), defined as an increase in either alanine aminotransferase (ALT), aspartate transaminase (AST), or bilirubin levels, was the only AE identified as a significant poor-prognosis indicator in this study. The median progression-free survival of patients with trHE (41.9%) was 8 months, whereas it was 18 months for those without trHE [p = 0.046, hazard ratio (HR) = 2.116]. Moreover, this association was independent of medication regimens (p = 0.014, HR = 2.971). The overall response rate of patients with trHE was significantly lower than those without trHE (44.4 vs. 60.0%, p = 0.024), and we observed a similar trend in patients treated with BRAFi, BRAFi+MEKi, and BRAFi+anti-PD-1 antibody. In conclusion, BRAFi and BRAFi-based combination therapies were tolerable with reversible AEs in Chinese patients with melanoma. The trHE in patients receiving BRAFi and BRAFi-based regimens might indicate a poor therapy-related prognosis.
Collapse
Affiliation(s)
- Xing Liu
- Biotherapy Center, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jing-Jing Li
- Biotherapy Center, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ya Ding
- Biotherapy Center, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Dan-Dan Li
- Biotherapy Center, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xi-Zhi Wen
- Biotherapy Center, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - De-Sheng Weng
- Biotherapy Center, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jiu-Hong Wang
- Biotherapy Center, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hang Jiang
- Biotherapy Center, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiao-Shi Zhang
- Biotherapy Center, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
621
|
Poizeau F, Kerbrat S, Happe A, Rault C, Drezen E, Balusson F, Tuppin P, Guillot B, Thuret A, Boussemart L, Dinulescu M, Pracht M, Lesimple T, Droitcourt C, Oger E, Dupuy A. Patients with Metastatic Melanoma Receiving Anticancer Drugs: Changes in Overall Survival, 2010–2017. J Invest Dermatol 2021; 141:830-839.e3. [DOI: 10.1016/j.jid.2020.07.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/30/2020] [Accepted: 07/13/2020] [Indexed: 12/19/2022]
|
622
|
Moosavi F, Giovannetti E, Peters GJ, Firuzi O. Combination of HGF/MET-targeting agents and other therapeutic strategies in cancer. Crit Rev Oncol Hematol 2021; 160:103234. [PMID: 33497758 DOI: 10.1016/j.critrevonc.2021.103234] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 12/29/2020] [Accepted: 01/16/2021] [Indexed: 02/06/2023] Open
Abstract
MET receptor has emerged as a druggable target across several human cancers. Agents targeting MET and its ligand hepatocyte growth factor (HGF) including small molecules such as crizotinib, tivantinib and cabozantinib or antibodies including rilotumumab and onartuzumab have proven their values in different tumors. Recently, capmatinib was approved for treatment of metastatic lung cancer with MET exon 14 skipping. In this review, we critically examine the current evidence on how HGF/MET combination therapies may take advantage of synergistic effects, overcome primary or acquired drug resistance, target tumor microenvironment, modulate drug metabolism or tackle pharmacokinetic issues. Preclinical and clinical studies on the combination of HGF/MET-targeted agents with conventional chemotherapeutics or molecularly targeted treatments (including EGFR, VEGFR, HER2, RAF/MEK, and PI3K/Akt targeting agents) and also the value of biomarkers are examined. Our deeper understanding of molecular mechanisms underlying successful pharmacological combinations is crucial to find the best personalized treatment regimens for cancer patients.
Collapse
Affiliation(s)
- Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), Amsterdam, the Netherlands; Cancer Pharmacology Lab, AIRC Start Up Unit, Fondazione Pisana per la Scienza, Pisa, Italy
| | - Godefridus J Peters
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), Amsterdam, the Netherlands; Department of Biochemistry, Medical University of Gdansk, Gdansk, Poland
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
623
|
Manzari MT, Shamay Y, Kiguchi H, Rosen N, Scaltriti M, Heller DA. Targeted drug delivery strategies for precision medicines. NATURE REVIEWS. MATERIALS 2021; 6:351-370. [PMID: 34950512 PMCID: PMC8691416 DOI: 10.1038/s41578-020-00269-6] [Citation(s) in RCA: 485] [Impact Index Per Article: 121.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/24/2020] [Indexed: 05/05/2023]
Abstract
Progress in the field of precision medicine has changed the landscape of cancer therapy. Precision medicine is propelled by technologies that enable molecular profiling, genomic analysis, and optimized drug design to tailor treatments for individual patients. Although precision medicines have resulted in some clinical successes, the use of many potential therapeutics has been hindered by pharmacological issues, including toxicities and drug resistance. Drug delivery materials and approaches have now advanced to a point where they can enable the modulation of a drug's pharmacological parameters without compromising the desired effect on molecular targets. Specifically, they can modulate a drug's pharmacokinetics, stability, absorption, and exposure to tumours and healthy tissues, and facilitate the administration of synergistic drug combinations. This Review highlights recent progress in precision therapeutics and drug delivery, and identifies opportunities for strategies to improve the therapeutic index of cancer drugs, and consequently, clinical outcomes.
Collapse
Affiliation(s)
- Mandana T. Manzari
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- These authors have contributed equally to this work
| | - Yosi Shamay
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
- These authors have contributed equally to this work
| | - Hiroto Kiguchi
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- These authors have contributed equally to this work
| | - Neal Rosen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Maurizio Scaltriti
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer, New York, NY, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel A. Heller
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
624
|
Bong CY, Smithers BM, Chua TC. Pulmonary metastasectomy in the era of targeted therapy and immunotherapy. J Thorac Dis 2021; 13:2618-2627. [PMID: 34012610 PMCID: PMC8107521 DOI: 10.21037/jtd.2020.03.120] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Metastatic melanoma is a fatal malignancy with a high mortality and morbidity. Since the early 1970s, available medical therapies were limited in improving survival. Surgery represented the best chance for a cure. However, surgery could only be offered to selected patients. The current landscape of treatment has radically evolved since the introduction of targeted and immunotherapies including BRAF and MEK inhibitors, and checkpoint blockers, like PD-1 and CTLA-4 antibodies. These new therapies have seen survival rates matching, and in some cases surpassing, that of surgery. Anti-PD1 and CTLA-4 combination treatments are associated with severe side effects and BRAF and MEK inhibitor combinations may trigger initial tumour responses but prolonged use have resulted in the development of resistant tumour clones and disease relapse. This review examines the role of pulmonary metastasectomy for lung metastasis from malignant melanoma in the current landscape of effective targeted therapy and immunotherapy.
Collapse
Affiliation(s)
- Christopher Y Bong
- Department of Surgery, Logan Hospital, Metro South Health, Meadowbrook, Queensland, Australia
| | - B Mark Smithers
- Upper Gastrointestinal and Soft Tissue Unit, Department of Surgery, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,Discipline of Surgery, Faculty of Medicine, The University of Queensland, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Terence C Chua
- Department of Surgery, Logan Hospital, Metro South Health, Meadowbrook, Queensland, Australia.,Discipline of Surgery, Faculty of Medicine, The University of Queensland, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,School of Medicine, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
625
|
Bourque MS, Salek M, Sabin ND, Canale M, Upadhyaya SA. Comment on: Response to the BRAF/MEK inhibitors dabrafenib/trametinib in an adolescent with a BRAF V600E mutated anaplastic ganglioglioma intolerant to vemurafenib. Pediatr Blood Cancer 2021; 68:e28814. [PMID: 33211390 DOI: 10.1002/pbc.28814] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 11/02/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Melissa S Bourque
- Department of Pharmaceutical Services, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Marta Salek
- Department of Hematology/Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Noah D Sabin
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Meredith Canale
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Santhosh A Upadhyaya
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
626
|
Abstract
The proliferation of targeted anticancer agents over the last two decades has revolutionized cancer treatment and improved survival in many previously refractory malignancies. However, many agents are associated with characteristic ophthalmic adverse effects. It is important that ophthalmologists recognize and maintain a high index of suspicion for these side effects in patients on targeted therapy. Most ophthalmic adverse effects can be treated with specific ocular therapy without discontinuation of cancer treatment, although it is important to be aware of the life-threatening and vision-threatening circumstances that would require therapy cessation in conjunction with the patient's oncologist. This review aims to summarize the ophthalmic adverse effects of targeted and hormonal anticancer agents and briefly describe their management.
Collapse
|
627
|
Sibaud V, Baric L, Cantagrel A, Di Palma M, Ederhy S, Paques M, Perlemuter G. [Management of toxicities of BRAF inhibitors and MEK inhibitors in advanced melanoma]. Bull Cancer 2021; 108:528-543. [PMID: 33812673 DOI: 10.1016/j.bulcan.2020.12.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/17/2020] [Accepted: 12/24/2020] [Indexed: 01/16/2023]
Abstract
Major therapeutic advances have been made recently in the treatment of metastatic melanoma, due to the development of targeted therapies, namely BRAF and MEK inhibitors, in patients with BRAF V600 mutation. Combinations of vemurafenib+cobimetinib, dabrafenib+trametinib, et encorafenib+binimetinib, evaluated in coBRIM, COMBI-d/COMBI-v and COLUMBUS trials respectively have been approved in this indication. Toxicities induced by combination therapies are different from those reported with monotherapies, in terms of frequency and intensity. Physicians who treat melanoma patients thus face news issues relating to prevention, detection and treatment of these adverse events. This paper summarizes tolerance data from the three pivotal trials (coBRIM, COMBI-v and COLUMBUS) and issues recommendations for the specific management of main toxicities, based on experts' opinion. We discuss dermatological, ophthalmological, cardiovascular, digestive, musculoskeletal, renal and general toxicities and propose a timetable for examinations to be performed before and during treatment.
Collapse
Affiliation(s)
- Vincent Sibaud
- Institut universitaire du cancer Toulouse - Oncopole, départements d'oncologie médicale et oncodermatologie, Toulouse, France.
| | - Lilian Baric
- Institut universitaire du cancer Toulouse - Oncopole, départements d'oncologie médicale et oncodermatologie, Toulouse, France
| | - Alain Cantagrel
- Hôpital Pierre-Paul-Riquet, CHU de Purpan, centre de rhumatologie, Toulouse, France
| | - Mario Di Palma
- Hôpital américain de Paris, service d'oncologie médicale, Neuilly-sur-Seine, France
| | - Stéphane Ederhy
- AP-HP, hôpital Saint-Antoine, service de cardiologie, unité de cardio-oncologie, groupe de recherche clinique en cardio-oncologie, Paris, France
| | - Michel Paques
- Sorbonne Université, centre hospitalier national d'ophtalmologie des Quinze-Vingts, Paris, France
| | - Gabriel Perlemuter
- AP-HP, hôpital Antoine-Béclère, service d'hépato-gastroentérologie et nutrition, 92140 Clamart, France; Université Paris-Saclay, inflammation, microbiome and immunosurveillance, Inserm U996, 92140 Clamart, France
| |
Collapse
|
628
|
Van Herck Y, Antoranz A, Andhari MD, Milli G, Bechter O, De Smet F, Bosisio FM. Multiplexed Immunohistochemistry and Digital Pathology as the Foundation for Next-Generation Pathology in Melanoma: Methodological Comparison and Future Clinical Applications. Front Oncol 2021; 11:636681. [PMID: 33854972 PMCID: PMC8040928 DOI: 10.3389/fonc.2021.636681] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/12/2021] [Indexed: 12/14/2022] Open
Abstract
The state-of-the-art for melanoma treatment has recently witnessed an enormous revolution, evolving from a chemotherapeutic, "one-drug-for-all" approach, to a tailored molecular- and immunological-based approach with the potential to make personalized therapy a reality. Nevertheless, methods still have to improve a lot before these can reliably characterize all the tumoral features that make each patient unique. While the clinical introduction of next-generation sequencing has made it possible to match mutational profiles to specific targeted therapies, improving response rates to immunotherapy will similarly require a deep understanding of the immune microenvironment and the specific contribution of each component in a patient-specific way. Recent advancements in artificial intelligence and single-cell profiling of resected tumor samples are paving the way for this challenging task. In this review, we provide an overview of the state-of-the-art in artificial intelligence and multiplexed immunohistochemistry in pathology, and how these bear the potential to improve diagnostics and therapy matching in melanoma. A major asset of in-situ single-cell profiling methods is that these preserve the spatial distribution of the cells in the tissue, allowing researchers to not only determine the cellular composition of the tumoral microenvironment, but also study tissue sociology, making inferences about specific cell-cell interactions and visualizing distinctive cellular architectures - all features that have an impact on anti-tumoral response rates. Despite the many advantages, the introduction of these approaches requires the digitization of tissue slides and the development of standardized analysis pipelines which pose substantial challenges that need to be addressed before these can enter clinical routine.
Collapse
Affiliation(s)
| | - Asier Antoranz
- Laboratory for Translational Cell and Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Madhavi Dipak Andhari
- Laboratory for Translational Cell and Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Giorgia Milli
- Laboratory for Translational Cell and Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | | | - Frederik De Smet
- Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Francesca Maria Bosisio
- Laboratory for Translational Cell and Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| |
Collapse
|
629
|
Clinical outcome of patients with metastatic melanoma of unknown primary in the era of novel therapy. Cancer Immunol Immunother 2021; 70:3123-3135. [PMID: 33774697 PMCID: PMC8505371 DOI: 10.1007/s00262-021-02871-1] [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: 11/23/2020] [Accepted: 01/20/2021] [Indexed: 11/25/2022]
Abstract
Melanoma of unknown primary (MUP) is considered different from melanoma of known primary (MKP), and it is unclear whether these patients benefit equally from novel therapies. In the current study, characteristics and overall survival (OS) of patients with advanced and metastatic MUP and MKP were compared in the era of novel therapy. Patients were selected from the prospective nation-wide Dutch Melanoma Treatment Registry (DMTR). The following criteria were applied: diagnosis of stage IIIc unresectable or IV cutaneous MKP (cMKP) or MUP between July 2012 and July 2017 and treatment with immune checkpoint inhibition and/or targeted therapy. OS was estimated using the Kaplan–Meier method. The stratified multivariable Cox regression model was used for adjusted analysis. A total of 2706 patients were eligible including 2321 (85.8%) patients with cMKP and 385 (14.2%) with MUP. In comparative analysis, MUP patients more often presented with advanced and metastatic disease at primary diagnosis with poorer performance status, higher LDH, and central nervous system metastases. In crude analysis, median OS of cMKP or MUP patients was 12 months (interquartile range [IQR] 5 – 44) and 14 months (IQR 5 – not reached), respectively (P = 0.278). In adjusted analysis, OS in MUP patients was superior (hazard rate 0.70, 95% confidence interval 0.58–0.85; P < 0.001). As compared to patients with advanced and metastatic cMKP, MUP patients have superior survival in adjusted analysis, but usually present with poorer prognostic characteristics. In crude analysis, OS was comparable indicating that patients with MUP benefit at least equally from treatment with novel therapies.
Collapse
|
630
|
Osrodek M, Wozniak M. Targeting Genome Stability in Melanoma-A New Approach to an Old Field. Int J Mol Sci 2021; 22:3485. [PMID: 33800547 PMCID: PMC8036881 DOI: 10.3390/ijms22073485] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Despite recent groundbreaking advances in the treatment of cutaneous melanoma, it remains one of the most treatment-resistant malignancies. Due to resistance to conventional chemotherapy, the therapeutic focus has shifted away from aiming at melanoma genome stability in favor of molecularly targeted therapies. Inhibitors of the RAS/RAF/MEK/ERK (MAPK) pathway significantly slow disease progression. However, long-term clinical benefit is rare due to rapid development of drug resistance. In contrast, immune checkpoint inhibitors provide exceptionally durable responses, but only in a limited number of patients. It has been increasingly recognized that melanoma cells rely on efficient DNA repair for survival upon drug treatment, and that genome instability increases the efficacy of both MAPK inhibitors and immunotherapy. In this review, we discuss recent developments in the field of melanoma research which indicate that targeting genome stability of melanoma cells may serve as a powerful strategy to maximize the efficacy of currently available therapeutics.
Collapse
Affiliation(s)
| | - Michal Wozniak
- Department of Molecular Biology of Cancer, Medical University of Lodz, 92-215 Lodz, Poland;
| |
Collapse
|
631
|
Ottaviano M, Giunta EF, Tortora M, Curvietto M, Attademo L, Bosso D, Cardalesi C, Rosanova M, De Placido P, Pietroluongo E, Riccio V, Mucci B, Parola S, Vitale MG, Palmieri G, Daniele B, Simeone E, on behalf of SCITO YOUTH. BRAF Gene and Melanoma: Back to the Future. Int J Mol Sci 2021; 22:ijms22073474. [PMID: 33801689 PMCID: PMC8037827 DOI: 10.3390/ijms22073474] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/20/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022] Open
Abstract
As widely acknowledged, 40-50% of all melanoma patients harbour an activating BRAF mutation (mostly BRAF V600E). The identification of the RAS-RAF-MEK-ERK (MAP kinase) signalling pathway and its targeting has represented a valuable milestone for the advanced and, more recently, for the completely resected stage III and IV melanoma therapy management. However, despite progress in BRAF-mutant melanoma treatment, the two different approaches approved so far for metastatic disease, immunotherapy and BRAF+MEK inhibitors, allow a 5-year survival of no more than 60%, and most patients relapse during treatment due to acquired mechanisms of resistance. Deep insight into BRAF gene biology is fundamental to describe the acquired resistance mechanisms (primary and secondary) and to understand the molecular pathways that are now being investigated in preclinical and clinical studies with the aim of improving outcomes in BRAF-mutant patients.
Collapse
Affiliation(s)
- Margaret Ottaviano
- Department of Clinical Medicine and Surgery, Università Degli Studi di Napoli “Federico II”, 80131 Naples, Italy; (P.D.P.); (E.P.); (V.R.); (B.M.); (S.P.)
- Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (L.A.); (D.B.); (C.C.); (M.R.); (B.D.)
- CRCTR Coordinating Rare Tumors Reference Center of Campania Region, 80131 Naples, Italy; (M.T.); (G.P.)
- Correspondence:
| | - Emilio Francesco Giunta
- Department of Precision Medicine, Università Degli Studi della Campania Luigi Vanvitelli, 80131 Naples, Italy;
| | - Marianna Tortora
- CRCTR Coordinating Rare Tumors Reference Center of Campania Region, 80131 Naples, Italy; (M.T.); (G.P.)
| | - Marcello Curvietto
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, 80131 Naples, Italy; (M.C.); (M.G.V.); (E.S.)
| | - Laura Attademo
- Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (L.A.); (D.B.); (C.C.); (M.R.); (B.D.)
| | - Davide Bosso
- Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (L.A.); (D.B.); (C.C.); (M.R.); (B.D.)
| | - Cinzia Cardalesi
- Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (L.A.); (D.B.); (C.C.); (M.R.); (B.D.)
| | - Mario Rosanova
- Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (L.A.); (D.B.); (C.C.); (M.R.); (B.D.)
| | - Pietro De Placido
- Department of Clinical Medicine and Surgery, Università Degli Studi di Napoli “Federico II”, 80131 Naples, Italy; (P.D.P.); (E.P.); (V.R.); (B.M.); (S.P.)
| | - Erica Pietroluongo
- Department of Clinical Medicine and Surgery, Università Degli Studi di Napoli “Federico II”, 80131 Naples, Italy; (P.D.P.); (E.P.); (V.R.); (B.M.); (S.P.)
| | - Vittorio Riccio
- Department of Clinical Medicine and Surgery, Università Degli Studi di Napoli “Federico II”, 80131 Naples, Italy; (P.D.P.); (E.P.); (V.R.); (B.M.); (S.P.)
| | - Brigitta Mucci
- Department of Clinical Medicine and Surgery, Università Degli Studi di Napoli “Federico II”, 80131 Naples, Italy; (P.D.P.); (E.P.); (V.R.); (B.M.); (S.P.)
| | - Sara Parola
- Department of Clinical Medicine and Surgery, Università Degli Studi di Napoli “Federico II”, 80131 Naples, Italy; (P.D.P.); (E.P.); (V.R.); (B.M.); (S.P.)
| | - Maria Grazia Vitale
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, 80131 Naples, Italy; (M.C.); (M.G.V.); (E.S.)
| | - Giovannella Palmieri
- CRCTR Coordinating Rare Tumors Reference Center of Campania Region, 80131 Naples, Italy; (M.T.); (G.P.)
| | - Bruno Daniele
- Oncology Unit, Ospedale del Mare, 80147 Naples, Italy; (L.A.); (D.B.); (C.C.); (M.R.); (B.D.)
| | - Ester Simeone
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, 80131 Naples, Italy; (M.C.); (M.G.V.); (E.S.)
| | | |
Collapse
|
632
|
Kiniwa Y, Okuyama R. Recent advances in molecular targeted therapy for unresectable and metastatic BRAF-mutated melanoma. Jpn J Clin Oncol 2021; 51:315-320. [PMID: 33338202 DOI: 10.1093/jjco/hyaa222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/21/2020] [Indexed: 12/15/2022] Open
Abstract
The clinical outcome of BRAF-mutated advanced melanoma has been improved by both molecular targeted therapies and immune checkpoint inhibitors. Long-term follow-up data reveal durable clinical responses in patients receiving first-line combinations of BRAF inhibitors plus MEK inhibitors, particularly those showing a complete response. Clinical outcomes are also associated with the lactate dehydrogenase levels and the number of metastatic organs. Although brain metastasis is frequently difficult to control, systemic therapy is preferred in cases with small and asymptomatic brain metastases associated with progressive extra-cranial disease. Control of intra-cranial disease with BRAF inhibitors plus MEK inhibitors is comparable with that of immune checkpoint inhibitors, although immune checkpoint inhibitors are superior to targeted therapies with respect to survival. The BRAF inhibitors plus MEK inhibitors regimen is well-tolerated, and toxicities are usually manageable and reversible, but differ according to the specific regimen used. Guidelines in the United States, Europe, and Japan recommend targeted therapy for patients who need early tumor responses. A meta-analysis of retrospective data shows that the baseline lactate dehydrogenase level is significantly higher in patients treated with BRAF inhibitors plus MEK inhibitors than in those treated with immune checkpoint inhibitors, suggesting that clinicians tend to use BRAF inhibitors plus MEK inhibitors for more advanced disease. Since there is insufficient efficacy and safety data on the use of targeted therapies for acral and mucosal melanoma, a retrospective analysis may be useful. The combination of molecular targeted therapy plus immune checkpoint inhibitors is expected to elicit further improvement. The results of several trials using combination or sequential therapies will be available in the next few years.
Collapse
Affiliation(s)
- Yukiko Kiniwa
- Department of Dermatology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Ryuhei Okuyama
- Department of Dermatology, Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
633
|
Cui H, Wang Q, Miller DD, Li W. The Tubulin Inhibitor VERU-111 in Combination With Vemurafenib Provides an Effective Treatment of Vemurafenib-Resistant A375 Melanoma. Front Pharmacol 2021; 12:637098. [PMID: 33841154 PMCID: PMC8027488 DOI: 10.3389/fphar.2021.637098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/08/2021] [Indexed: 11/13/2022] Open
Abstract
Melanoma is one of the deadliest skin cancers having a five-year survival rate around 15–20%. An overactivated MAPK/AKT pathway is well-established in BRAF mutant melanoma. Vemurafenib (Vem) was the first FDA-approved BRAF inhibitor and gained great clinical success in treating late-stage melanoma. However, most patients develop acquired resistance to Vem within 6–9 months. Therefore, developing a new treatment strategy to overcome Vem-resistance is highly significant. Our previous study reported that the combination of a tubulin inhibitor ABI-274 with Vem showed a significant synergistic effect to sensitize Vem-resistant melanoma both in vitro and in vivo. In the present study, we unveiled that VERU-111, an orally bioavailable inhibitor of α and β tubulin that is under clinical development, is highly potent against Vem-resistant melanoma cells. The combination of Vem and VERU-111 resulted in a dramatically enhanced inhibitory effect on cancer cells in vitro and Vem-resistant melanoma tumor growth in vivo compared with single-agent treatment. Further molecular signaling analyses demonstrated that in addition to ERK/AKT pathway, Skp2 E3 ligase also plays a critical role in Vem-resistant mechanisms. Knockout of Skp2 diminished oncogene AKT expression and contributed to the synergistic inhibitory effect of Vem and VERU-111. Our results indicate a treatment combination of VERU-111 and Vem holds a great promise to overcome Vem-resistance for melanoma patients harboring BRAF (V600E) mutation.
Collapse
Affiliation(s)
- Hongmei Cui
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, United States.,Institute of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Qinghui Wang
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Duane D Miller
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Wei Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
634
|
Abstract
The management of melanoma significantly improved within the last 25 years. Chemotherapy was the first approved systemic therapeutic approach and resulted in a median overall of survival less than 1 year, without survival improvement in phase III trials. High-dose interferon α2b and IL-2 were introduced for resectable high-risk and advanced disease, respectively, resulting in improved survival and response rates. The anti-CTLA4 and anti-programmed death 1 monoclonal antibodies along with BRAF/MEK targeted therapies are the dominant therapeutic classes of agent for melanoma. This article provides an historic overview of the evolution of melanoma management.
Collapse
|
635
|
Steininger J, Gellrich FF, Schulz A, Westphal D, Beissert S, Meier F. Systemic Therapy of Metastatic Melanoma: On the Road to Cure. Cancers (Basel) 2021; 13:1430. [PMID: 33804800 PMCID: PMC8003858 DOI: 10.3390/cancers13061430] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 12/22/2022] Open
Abstract
This decade has brought significant survival improvement in patients with metastatic melanoma with targeted therapies and immunotherapies. As our understanding of the mechanisms of action of these therapeutics evolves, even more impressive therapeutic success is being achieved through various combination strategies, including combinations of different immunotherapies as well as with other modalities. This review summarizes prospectively and retrospectively generated clinical evidence on modern melanoma therapy, focusing on immunotherapy and targeted therapy with BRAF kinase inhibitors and MEK kinase inhibitors (BRAF/MEK inhibitors), including recent data presented at major conference meetings. The combination of the anti-PD-1 directed monoclonal antibody nivolumab and of the CTLA-4 antagonist ipilimumab achieves unprecedented 5-year overall survival (OS) rates above 50%; however, toxicity is high. For PD-1 monotherapy (nivolumab or pembrolizumab), toxicities are in general well manageable. Today, novel combinations of such immune checkpoint inhibitors (ICIs) are under investigation, for example with cytokines and oncolytic viruses (i.e., pegylated interleukin-2, talimogene laherparepvec). Furthermore, current studies investigate the combined or sequential use of ICIs plus BRAF/MEK inhibitors. Several studies focus particularly on poor prognosis patients, as e.g., on anti-PD-1 refractory melanoma, patients with brain metastases, or uveal melanoma. It is hoped, on the road to cure, that these new approaches further improve long term survival in patients with advanced or metastatic melanoma.
Collapse
|
636
|
Vanella V, Festino L, Vitale MG, Alfano B, Ascierto PA. Emerging PD-1/PD-L1 antagonists for the treatment of malignant melanoma. Expert Opin Emerg Drugs 2021; 26:79-92. [PMID: 33686894 DOI: 10.1080/14728214.2021.1901884] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Increased understanding of the interactive mechanisms between tumors and the immune system led to the development of immune checkpoint inhibitors, which have revolutioned the treatment of metastatic melanoma and subsequently many other tumors. In 2014, nivolumab and pembrolizumab, two checkpoint inhibitors binding to PD-1, were approved for the treatment of metastatic melanoma. Since then, a plethora of new molecules have enriched the armamentarium against melanoma. AREAS COVERED This review summarizes the last updates about treatment with nivolumab and pembrolizumab, data on other PD-1/PDL-1 agents such as spartalizumab and atezolizumab and emerging compounds, new combinations with NKTR-214, anti LAG-3, anti IDO-1 and TVEC, new checkpoint inhibitors (e.g. TIM-3 or TIGIT) and other new molecules for the treatment of metastatic melanoma. EXPERT OPINION Currently, several ongoing clinical trials are investigating novel molecules, or immunotherapy combinations, in order to achieve even better survival outcomes for patients, overcoming resistance mechanisms and improving toxicity profiles. The challenge in the near future will be to select the most appropriate treatments according to the specific characteristics of the patients.
Collapse
Affiliation(s)
- Vito Vanella
- Melanoma Unit, Cancer Immunotherapy and Innovative Therapies, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| | - Lucia Festino
- Melanoma Unit, Cancer Immunotherapy and Innovative Therapies, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| | - Maria Grazia Vitale
- Melanoma Unit, Cancer Immunotherapy and Innovative Therapies, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| | - Benedetta Alfano
- Melanoma Unit, Cancer Immunotherapy and Innovative Therapies, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| | - Paolo Antonio Ascierto
- Melanoma Unit, Cancer Immunotherapy and Innovative Therapies, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| |
Collapse
|
637
|
Knispel S, Gassenmaier M, Menzies AM, Loquai C, Johnson DB, Franklin C, Gutzmer R, Hassel JC, Weishaupt C, Eigentler T, Schilling B, Schummer P, Sirokay J, Kiecker F, Owen CN, Fleischer MI, Cann C, Kähler KC, Mohr P, Bluhm L, Niebel D, Thoms KM, Goldinger SM, Reinhardt L, Meier F, Berking C, Reinhard R, Susok L, Ascierto PA, Drexler K, Pföhler C, Tietze J, Heinzerling L, Livingstone E, Ugurel S, Long GV, Stang A, Schadendorf D, Zimmer L. Outcome of melanoma patients with elevated LDH treated with first-line targeted therapy or PD-1-based immune checkpoint inhibition. Eur J Cancer 2021; 148:61-75. [PMID: 33735811 DOI: 10.1016/j.ejca.2021.01.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/24/2021] [Accepted: 01/28/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Elevated lactate dehydrogenase (LDH) is a known predictive and prognostic factor for a poor outcome in patients with metastatic melanoma. It is unclear whether first-line targeted therapy (TT) or immune checkpoint inhibition (ICI) is more beneficial in melanoma patients with elevated LDH because prospective studies in this area are lacking. METHODS This multicentre retrospective cohort study was conducted at 25 melanoma centres worldwide to analyse progression-free survival (PFS) and overall survival (OS) among melanoma patients with elevated LDH. The role of confounders was addressed by using inverse probability of treatment weighting. RESULTS Among 173 BRAFV600-mutant patients, PFS at 12 months in the TT group was 22% compared with 52% in the combined anti-PD-1 and anti-CTLA-4 group (HR 0.6, 95% CI 0.4-1.0, p = 0.07) and 18% in the anti-PD-1 monotherapy group (HR 1.8, 95% CI 1.2-2.8, p = 0.003). Twelve months' OS was 48% in the TT group compared with 83% in the combined anti-PD-1 and anti-CTLA-4 group (HR 0.5, 95% CI 0.3-1.0, p = 0.03) and 50% in the anti-PD-1 monotherapy group (HR 1.2, 95% CI 0.8-2.0, p = 0.37). The ORR in the TT group was 63%, compared with 55% and 20% in the combined anti-PD-1 and anti-CTLA-4 and anti-PD-1 monotherapy group, respectively. Among 314 patients receiving ICI first-line, PFS at 12 months was 33% in the anti-PD-1 group versus 38% in the combined anti-PD-1 and anti-CTLA-4 group (HR 0.8, 95% CI 0.6-1.0; p = 0.07). OS at 12 months was 54% in the anti-PD-1 group versus 66% in the combined ICI group (HR 0.7, 95% CI 0.5-1.0; p = 0.03). The ORR was 30% in the anti-PD-1 monotherapy group and 43% in the combined anti-PD-1 and anti-CTLA-4 group. Results from multivariate analysis confirmed the absence of qualitative confounding. CONCLUSIONS Among BRAF-mutant patients with elevated LDH, combined anti-PD-1 and anti-CTLA-4 blockade seems to be associated with prolonged OS compared with first-line TT. Among patients receiving ICI as a first-line treatment, OS appears to be longer for the combination of anti-PD-1 and anti-CTLA-4 than for anti-PD-1 alone.
Collapse
Affiliation(s)
- Sarah Knispel
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | | | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Department of Medical Oncology, Royal North Shore and Mater Hospitals, Sydney, Australia
| | - Carmen Loquai
- Department of Dermatology, University Medical Center Mainz, Mainz, Germany
| | | | - Cindy Franklin
- Department of Dermatology and Venereology, Skin Cancer Center at the Center of Integrated Oncology (CIO) Köln Bonn, University Hospital of Cologne, Cologne, Germany
| | - Ralf Gutzmer
- Department of Dermatology and Allergy, Skin Cancer Center Hannover, Hannover Medical School, Hannover, Germany
| | - Jessica C Hassel
- Skin Cancer Center, Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Carsten Weishaupt
- Department of Dermatology, University Hospital of Muenster, Muenster, Germany
| | - Thomas Eigentler
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Bastian Schilling
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - Patrick Schummer
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - Judith Sirokay
- Department of Dermatology, University Hospital Bonn, Bonn, Germany
| | - Felix Kiecker
- Department of Dermatology, University Hospital Charité Berlin, Berlin, Germany
| | - Carina N Owen
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia
| | - Maria I Fleischer
- Department of Dermatology, University Medical Center Mainz, Mainz, Germany
| | | | - Katharina C Kähler
- Department of Dermatology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Peter Mohr
- Department of Dermatology, Elbe-Klinikum Buxtehude, Buxtehude, Germany
| | - Leonie Bluhm
- Department of Dermatology, Elbe-Klinikum Buxtehude, Buxtehude, Germany
| | - Dennis Niebel
- Department of Dermatology, University Hospital Bonn, Bonn, Germany
| | - Kai-Martin Thoms
- Department of Dermatology, University Medical Center Goettingen, Göttingen, Germany
| | - Simone M Goldinger
- Department of Dermatology, University Hospital Zürich, Zürich, Switzerland
| | - Lydia Reinhardt
- Department of Dermatology, Skin Cancer Center at the National Center for Tumor Diseases, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Friedegund Meier
- Department of Dermatology, Skin Cancer Center at the National Center for Tumor Diseases, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Carola Berking
- Department of Dermatology, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen - Metropolitan Region of Nuremberg, Deutsches Zentrum Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Raphael Reinhard
- Skin Cancer Unit, German Cancer Research Center (DKFZ) and Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Mannheim, Germany
| | - Laura Susok
- Department of Dermatology, St. Josef-Hospital Bochum, Bochum, Germany
| | - Paolo A Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy
| | - Konstantin Drexler
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Claudia Pföhler
- Department of Dermatology, Saarland University Medical School, Homburg/Saar, Germany
| | - Julia Tietze
- Department of Dermatology, University Hospital Rostock, Rostock, Germany
| | - Lucie Heinzerling
- Department of Dermatology, Universitätsklinikum Erlangen, Comprehensive Cancer Center Erlangen - Metropolitan Region of Nuremberg, Deutsches Zentrum Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Elisabeth Livingstone
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Department of Medical Oncology, Royal North Shore and Mater Hospitals, Sydney, Australia
| | - Andreas Stang
- Institute for Medical Informatics, Biometry and Epidemiology, University of Duisburg-Essen, Essen, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
638
|
Jacobs C, Lapeire L. Translating Molecular Profiling of Soft Tissue Sarcomas into Daily Clinical Practice. Diagnostics (Basel) 2021; 11:diagnostics11030512. [PMID: 33799327 PMCID: PMC7999686 DOI: 10.3390/diagnostics11030512] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/05/2021] [Accepted: 03/06/2021] [Indexed: 12/22/2022] Open
Abstract
Soft tissue sarcomas are a group of rare mesenchymal tumors with more than 70 subtypes described. Treatment of these subtypes in an advanced setting is mainly according to a one-size-fits-all strategy indicating a high unmet need of new and more targeted therapeutic options in order to optimize survival. The introduction of advanced molecular techniques in cancer has led to better diagnostics and identification of new therapeutic targets, leading to more personalized treatment and improved prognosis for several cancer types. In sarcoma, a likewise evolution is seen, albeit at a slower pace. This manuscript describes how in the past years advanced molecular profiling in soft tissue sarcomas was able to identify specific and often pathognomonic aberrations, deferring standard sarcoma treatment in favor of more targeted treatment from an oncologist’s point of view.
Collapse
Affiliation(s)
- Celine Jacobs
- Department of Medical Oncology, Ghent University Hospital, 9000 Ghent, Belgium;
| | - Lore Lapeire
- Department of Medical Oncology, Ghent University Hospital, 9000 Ghent, Belgium;
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium
- Correspondence:
| |
Collapse
|
639
|
Blood-Based Detection of BRAF V600E in Gliomas and Brain Tumor Metastasis. Cancers (Basel) 2021; 13:cancers13061227. [PMID: 33799709 PMCID: PMC7998685 DOI: 10.3390/cancers13061227] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 03/03/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary The BRAF V600E mutation has been identified as a key driver in brain tumors and brain tumor metastasis. The ability to detect this mutation in a minimally invasive plasma assay offers advantages over traditional tissue-based biopsy for the disease diagnosis and monitoring. The aim of this study was to develop an assay for the detection of BRAF V600E in the plasma of patients with brain tumors and brain tumor metastasis. We demonstrate BRAF V600E detection using a novel plasma-based ddPCR assay. We detect the mutation in circulating nucleic acids in 4/5 patients with mutant gliomas and metastatic melanoma. We also show correlation between plasma BRAF V600E and clinical status. This proof of principle study is important in the context of application of liquid biopsy in plasma to the neuro-oncologic field. The assay may be useful as a diagnostic adjunct, prognostication tool, and method for monitoring of disease and treatment response. Abstract Liquid biopsy provides a minimally invasive platform for the detection of tumor-derived information, including hotspot mutations, such as BRAF V600E. In this study, we provide evidence of the technical development of a ddPCR assay for the detection of BRAF V600E mutations in the plasma of patients with glioma or brain metastasis. In a small patient cohort (n = 9, n = 5 BRAF V600E, n = 4 BRAF WT, n = 4 healthy control), we were able to detect the BRAF V600E mutation in the plasma of 4/5 patients with BRAF V600E-tissue confirmed mutant tumors, and none of the BRAF WT tumors. We also provide evidence in two metastatic patients with longitudinal monitoring, where the plasma-based BRAF V600E mutation correlated with clinical disease status. This proof of principle study demonstrates the potential of this assay to serve as an adjunctive tool for the detection, monitoring, and molecular characterization of BRAF mutant gliomas and brain metastasis.
Collapse
|
640
|
Berta J, Török S, Tárnoki-Zách J, Drozdovszky O, Tóvári J, Paku S, Kovács I, Czirók A, Masri B, Megyesfalvi Z, Oskolás H, Malm J, Ingvar C, Markó-Varga G, Döme B, László V. Apelin promotes blood and lymph vessel formation and the growth of melanoma lung metastasis. Sci Rep 2021; 11:5798. [PMID: 33707612 PMCID: PMC7952702 DOI: 10.1038/s41598-021-85162-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/24/2021] [Indexed: 01/01/2023] Open
Abstract
Apelin, a ligand of the APJ receptor, is overexpressed in several human cancers and plays an important role in tumor angiogenesis and growth in various experimental systems. We investigated the role of apelin signaling in the malignant behavior of cutaneous melanoma. Murine B16 and human A375 melanoma cell lines were stably transfected with apelin encoding or control vectors. Apelin overexpression significantly increased melanoma cell migration and invasion in vitro, but it had no impact on its proliferation. In our in vivo experiments, apelin significantly increased the number and size of lung metastases of murine melanoma cells. Melanoma cell proliferation rates and lymph and blood microvessel densities were significantly higher in the apelin-overexpressing pulmonary metastases. APJ inhibition by the competitive APJ antagonist MM54 significantly attenuated the in vivo pro-tumorigenic effects of apelin. Additionally, we detected significantly elevated circulating apelin and VEGF levels in patients with melanoma compared to healthy controls. Our results show that apelin promotes blood and lymphatic vascularization and the growth of pulmonary metastases of skin melanoma. Further studies are warranted to validate apelin signaling as a new potential therapeutic target in this malignancy.
Collapse
Affiliation(s)
- Judit Berta
- Department of Tumor Biology, National Korányi Institute of Pulmonology, Budapest, Hungary
| | - Szilvia Török
- Department of Tumor Biology, National Korányi Institute of Pulmonology, Budapest, Hungary
| | | | - Orsolya Drozdovszky
- Department of Tumor Biology, National Korányi Institute of Pulmonology, Budapest, Hungary
| | - József Tóvári
- Department of Experimental Pharmacology, National Institute of Oncology, Budapest, Hungary
| | - Sándor Paku
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Ildikó Kovács
- Department of Tumor Biology, National Korányi Institute of Pulmonology, Budapest, Hungary
| | - András Czirók
- Department of Biological Physics, Eötvös University, Budapest, Hungary
- Department of Anatomy and Cell Biology, Medical Center, University of Kansas, Kansas City, KS, USA
- University of Kansas Cancer Center, Kansas City, KS, USA
| | - Bernard Masri
- Department of Endocrinology, Metabolism and Diabetes, Institute Cochin, INSERM U1016, CNRS UMR8104, Université de Paris, Paris, France
| | - Zsolt Megyesfalvi
- Department of Tumor Biology, National Korányi Institute of Pulmonology, Budapest, Hungary
- Translational Thoracic Oncology Laboratory, Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
- Department of Thoracic Surgery, National Institute of Oncology and Semmelweis University, Budapest, Hungary
| | - Henriett Oskolás
- Clinical Protein Science and Imaging, Biomedical Center, Department of Biomedical Engineering, Lund University, Lund, Sweden
| | - Johan Malm
- Department of Translational Medicine, Section for Clinical Chemistry, Lund University, Malmö, Sweden
| | | | - György Markó-Varga
- Clinical Protein Science and Imaging, Biomedical Center, Department of Biomedical Engineering, Lund University, Lund, Sweden
| | - Balázs Döme
- Department of Tumor Biology, National Korányi Institute of Pulmonology, Budapest, Hungary.
- Translational Thoracic Oncology Laboratory, Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria.
- Department of Thoracic Surgery, National Institute of Oncology and Semmelweis University, Budapest, Hungary.
| | - Viktória László
- Department of Tumor Biology, National Korányi Institute of Pulmonology, Budapest, Hungary.
- Translational Thoracic Oncology Laboratory, Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
641
|
A case of multi-metastatic melanoma with RAF1 fusion: a surprising response to anti-MEK therapy. Eur J Cancer 2021; 147:161-163. [PMID: 33684875 DOI: 10.1016/j.ejca.2021.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/29/2021] [Accepted: 02/03/2021] [Indexed: 01/23/2023]
|
642
|
Abstract
Although new compounds have improved the treatment landscape of metastatic melanoma, very limited data exist on the efficacy and safety of treating older patients with novel agents. Here, we provide results of BRAF (BRAFi) ± MEK (MEKi) inhibitor treatment in patients over 75 years (oldest-old patients) with metastatic melanoma. Between 2011 and 2020, 34 consecutive patients with metastatic melanoma over 75 years of age (range 75-89) were treated with BRAFi ± MEKi at the Comprehensive Cancer Center of Helsinki University Hospital. Data on clinical and histopathological features, toxicity, response rate (RR), progression-free survival (PFS) and overall survival (OS) were collected. Patients were treated with BRAFi (n = 22) or BRAFi in combination with MEK inhibitor (MEKi) (n = 12). Grade 1-2 adverse events occurred in 68% of the patients, 32% had grade 3 adverse effects, dose reductions were made for 41% of patients and 29% terminated treatment due to toxicity. Overall, the RR was 62%. Complete responses were achieved in 27% of the patients, and 35% had partial responses. The median PFS was 8 months (range 0-57), and the median OS was 15 months (range 0-71). Tailored BRAFi ± MEKi treatment for older patients is feasible. Adverse effects occur frequently but are manageable by dose adjustment. The occurrence of toxicity of monotherapy was similar to that of combination therapy. The RR and median OS from our retrospective study are comparable with those reported in clinical trials and combination therapy produced somewhat more and longer-lasting responses. Hence, it seems that older patients may benefit from BRAFi treatment.
Collapse
|
643
|
Salem D, Chelvanambi M, Storkus WJ, Fecek RJ. Cutaneous Melanoma: Mutational Status and Potential Links to Tertiary Lymphoid Structure Formation. Front Immunol 2021; 12:629519. [PMID: 33746966 PMCID: PMC7970117 DOI: 10.3389/fimmu.2021.629519] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 02/11/2021] [Indexed: 12/21/2022] Open
Abstract
Recent advances in immunotherapy have enabled rapid evolution of novel interventional approaches designed to reinvigorate and expand patient immune responses against cancer. An emerging approach in cancer immunology involves the conditional induction of tertiary lymphoid structures (TLS), which are non-encapsulated ectopic lymphoid structures forming at sites of chronic, pathologic inflammation. Cutaneous melanoma (CM), a highly-immunogenic form of solid cancer, continues to rise in both incidence and mortality rate, with recent reports supporting a positive correlation between the presence of TLS in melanoma and beneficial treatment outcomes amongst advanced-stage patients. In this context, TLS in CM are postulated to serve as dynamic centers for the initiation of robust anti-tumor responses within affected regions of active disease. Given their potential importance to patient outcome, significant effort has been recently devoted to gaining a better understanding of TLS neogenesis and the influence these lymphoid organs exert within the tumor microenvironment. Here, we briefly review TLS structure, function, and response to treatment in the setting of CM. To uncover potential tumor-intrinsic mechanisms that regulate TLS formation, we have taken the novel perspective of evaluating TLS induction in melanomas impacted by common driver mutations in BRAF, PTEN, NRAS, KIT, PRDM1, and MITF. Through analysis of The Cancer Genome Atlas (TCGA), we show expression of DNA repair proteins (DRPs) including BRCA1, PAXIP, ERCC1, ERCC2, ERCC3, MSH2, and PMS2 to be negatively correlated with expression of pro-TLS genes, suggesting DRP loss may favor TLS development in support of improved patient outcome and patient response to interventional immunotherapy.
Collapse
Affiliation(s)
- Deepak Salem
- Department of Microbiology, Lake Erie College of Osteopathic Medicine at Seton Hill, Greensburg, PA, United States
| | - Manoj Chelvanambi
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Ronald J Fecek
- Department of Microbiology, Lake Erie College of Osteopathic Medicine at Seton Hill, Greensburg, PA, United States
| |
Collapse
|
644
|
Clinical Significance of PDCD4 in Melanoma by Subcellular Expression and in Tumor-Associated Immune Cells. Cancers (Basel) 2021; 13:cancers13051049. [PMID: 33801444 PMCID: PMC7958624 DOI: 10.3390/cancers13051049] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/16/2021] [Accepted: 02/22/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary While targeting programmed cell death (PDCD) 1 is a central treatment against melanoma, little is known about the related protein PDCD4. We defined differences in melanoma PDCD4 subcellular localization (either total cellular or nuclear-only) during oncogenesis, evaluated its presence on tumor-infiltrating immune cells, and determined its impact on survival. High PDCD4 expression resulted in improved survival in patients with primary and intracranial but not extracranial metastatic melanoma. High PDCD4 levels in surrounding tumor tissue were also associated with increased infiltrating immune cells. PDCD4 may be a potentially useful biomarker in melanoma to help guide our understanding of patient prognosis. Methods to increase PDCD4 in those with melanoma brain metastases may also help improve disease response. Abstract Little is known about the subcellular localization and function of programmed cell death 4 (PDCD4) in melanoma. Our past studies suggest PDCD4 interacts with Pleckstrin Homology Domain Containing A5 (PLEKHA5) to influence melanoma brain metastasis outcomes, as high intracranial PDCD4 expression leads to improved survival. We aimed to define the subcellular distribution of PDCD4 in melanoma and in the tumor microenvironment during neoplastic progression and its impact on clinical outcomes. We analyzed multiple tissue microarrays with well-annotated clinicopathological variables using quantitative immunofluorescence and evaluated single-cell RNA-sequencing on a brain metastasis sample to characterize PDCD4+ immune cell subsets. We demonstrate differences in PDCD4 expression during neoplastic progression, with high tumor and stromal PDCD4 levels associated with improved survival in primary melanomas and in intracranial metastases, but not in extracranial metastatic disease. While the expression of PDCD4 is well-documented on CD8+ T cells and natural killer cells, we show that it is also found on B cells and mast cells. PDCD4 expression in the tumor microenvironment is associated with increased immune cell infiltration. Further studies are needed to define the interaction of PDCD4 and PLEKHA5 and to evaluate the utility of this pathway as a therapeutic target in melanoma brain metastasis.
Collapse
|
645
|
Gattozzi DA, Rosso C, Schatmeyer BA, Kabangu JLK, Doolittle GC, Wang F, Stepp T. Incidence of Intracranial Melanoma Progression in the Setting of Positive Extracranial Response to Targeted Therapy and Immunotherapy: An Indication for More Frequent Screening in This Population? Cureus 2021; 13:e13648. [PMID: 33824801 PMCID: PMC8013837 DOI: 10.7759/cureus.13648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background and objective The incidence of intracranial metastases from melanoma is on the rise. In this study, we aimed to determine the incidence of intracranial disease progression in patients on BRAF/MEK targeted therapy and immunotherapy in the setting of controlled or improving extracranial disease. Methods This was a single-center, retrospective review that involved patients who underwent stereotactic radiosurgery (SRS) for intracranial metastatic melanoma between January 1, 2014, and December 31, 2018. We focused on BRAF/MEK mutation status and dates of treatment with BRAF/MEK targeted therapy, immunotherapy [ipilimumab (Yervoy), nivolumab (Opdivo), or pembrolizumab (Keytruda)], and combination targeted and immunotherapy. Results A total of 51 patients were enrolled: 36 males and 15 females. The average age of the patients was 58.6 years, and 26 among them were BRAF mutation-positive. Seventeen had prior surgery with SRS as adjuvant therapy. The other 34 had SRS as primary treatment. Forty-two patients had extracranial disease present at the time of SRS. There were 34 patients treated with targeted and immune therapy. Overall, 16 patients (47.1%) demonstrated controlled or improving extracranial disease, and 18 (52.9%) demonstrated progressing extracranial disease at the time of SRS. In the subgroup analysis, patients treated with BRAF/MEK targeted therapy demonstrated a 75% rate of extracranial disease control. The extracranial disease was controlled in 43.75% of patients on immunotherapy with intracranial progression, while it was controlled in 30% of patients on both BRAF/MEK targeted therapy and immunotherapy with intracranial progression. Sixteen patients (47.1%) developed intracranial metastasis in our study while having a stable systemic disease with BRAF/MEK targeted therapy, immunotherapy, or a combination of the two. Conclusion Based on our findings, a systemic response to targeted therapy and immunotherapy does not necessarily parallel intracranial protection.
Collapse
Affiliation(s)
| | - Casey Rosso
- Neurosurgery, University of Kansas Medical School, Kansas City, USA
| | | | | | - Gary C Doolittle
- Hematology/Oncology, University of Kansas Medical Center, Kansas City, USA
| | - Fen Wang
- Radiation Oncology, University of Kansas Medical Center, Kansas City, USA
| | - Timothy Stepp
- Neurosurgery, University of Kansas Medical Center, Kansas City, USA
| |
Collapse
|
646
|
Cherri S, Noventa S, Fanelli M, Calandra G, Prochilo T, Bnà C, Savelli G, Zaniboni A. Drug-Related Pneumonitis in Cancer Treatment during the COVID-19 Era. Cancers (Basel) 2021; 13:1052. [PMID: 33801385 PMCID: PMC7958630 DOI: 10.3390/cancers13051052] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 02/24/2021] [Indexed: 12/17/2022] Open
Abstract
Interstitial lung disease is recognized as a group of diseases with a different etiopathogenesis characterized by chronic lung inflammation with the accumulation of inflammatory cells, lymphocytes and macrophages, and the consequent release of proinflammatory cytokines. Various degrees of pulmonary fibrosis can be associated with this inflammatory condition. Interstitial lung disease related to oncological drugs is a relevant problem in clinical practice. The etiopathogenetic mechanisms underlying this adverse event are not completely known but can be partly explained by the mechanism of action of the drug involved. Therefore, knowledge of the relevance of this potentially fatal adverse event supported by the reported safety data of pivotal studies becomes fundamental in the management of patients. The prompt diagnosis of drug-related pneumonia and the consequent differential diagnosis with other forms of pneumonia allow a rapid suspension of treatment and the establishment of an immunosuppressive treatment if necessary. In the context of the health emergency related to SARS CoV2 infection and COVID-19-related interstitial lung disease, such knowledge holds decisive relevance in the conscious choice of cancer treatments. Our intent was to describe the oncological drugs most correlated with this adverse event by reporting, where possible, the percentages of insurgency in pivotal studies to provide an overview and therefore promote greater awareness of this important toxicity related to oncological treatment.
Collapse
Affiliation(s)
- Sara Cherri
- Unit of Medical Oncology, Department of Oncology, Fondazione Poliambulanza, 25124 Brescia, Italy; (S.N.); (T.P.); (A.Z.)
| | - Silvia Noventa
- Unit of Medical Oncology, Department of Oncology, Fondazione Poliambulanza, 25124 Brescia, Italy; (S.N.); (T.P.); (A.Z.)
| | - Martina Fanelli
- Medical Oncology Unit, University Hospital of Modena, 41124 Modena, Italy;
| | - Giulio Calandra
- Unit of Radiology, Department of Diagnostic Imaging, Fondazione Poliambulanza, 25124 Brescia, Italy; (G.C.); (C.B.)
| | - Tiziana Prochilo
- Unit of Medical Oncology, Department of Oncology, Fondazione Poliambulanza, 25124 Brescia, Italy; (S.N.); (T.P.); (A.Z.)
| | - Claudio Bnà
- Unit of Radiology, Department of Diagnostic Imaging, Fondazione Poliambulanza, 25124 Brescia, Italy; (G.C.); (C.B.)
| | - Giordano Savelli
- Nuclear Medicine Department, Fondazione Poliambulanza, 25124 Brescia, Italy;
| | - Alberto Zaniboni
- Unit of Medical Oncology, Department of Oncology, Fondazione Poliambulanza, 25124 Brescia, Italy; (S.N.); (T.P.); (A.Z.)
| |
Collapse
|
647
|
Bander ED, Yuan M, Carnevale JA, Reiner AS, Panageas KS, Postow MA, Tabar V, Moss NS. Melanoma brain metastasis presentation, treatment, and outcomes in the age of targeted and immunotherapies. Cancer 2021; 127:2062-2073. [PMID: 33651913 DOI: 10.1002/cncr.33459] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 12/17/2020] [Accepted: 12/22/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Historically, the prognosis for patients who have melanoma brain metastasis (MBM) has been dismal. However, breakthroughs in targeted and immunotherapies have improved long-term survival in those with advanced melanoma. Therefore, MBM presentation, prognosis, and the use of multimodality central nervous system (CNS)-directed treatment were reassessed. METHODS In this retrospective study, the authors evaluated patients with MBM who received treatment at Memorial Sloan Kettering Cancer Center between 2010 and 2019. Kaplan-Meier methodology was used to describe overall survival (OS). Recursive partitioning analysis and time-dependent multivariable Cox modeling were used to assess prognostic variables and to associate CNS-directed treatments with OS. RESULTS Four hundred twenty-five patients with 2488 brain metastases were included. The median OS after an MBM diagnosis was 8.9 months (95% CI, 7.9-11.3 months). Patients who were diagnosed with MBM between 2015 and 2019 experienced longer OS compared to those who were diagnosed between 2010 and 2014 (OS, 13.0 months [95% CI, 10.47-17.06 months] vs 7.0 months [95% CI, 6.1-8.3 months]; P = .0003). Prognostic multivariable modeling significantly associated shortened OS independently with leptomeningeal dissemination (P < .0001), increasing numbers of brain metastases at diagnosis (P < .0001), earlier MBM diagnosis year (P = .0008), higher serum levels of lactate dehydrogenase (P < .0001), receipt of immunotherapy before MBM diagnosis (P = .003), and the presence of extracranial disease (P = .02). The use of different CNS-directed treatment modalities was associated with presenting symptoms, diagnosis year, number and size of brain metastases, and the presence of extracranial disease. Multivariable analysis demonstrated improved survival for patients who underwent craniotomy (P = .01). CONCLUSIONS The prognosis for patients with MBM has improved within the last 5 years, coinciding with the approval of PD-1 immune checkpoint blockade and combined BRAF/MEK targeting. Improving survival reflects and may influence the willingness to use aggressive multimodality treatment for MBM. LAY SUMMARY Historically, melanoma brain metastases (MBM) have carried a poor survival prognosis of 4 to 6 months; however, the introduction of immunotherapy and targeted precision medicines has altered the survival curve for advanced melanoma. In this large, single-institution, contemporary cohort, the authors demonstrate a significant increase in survival of patients with MBM to 13 months within the last 5 years of the study. A worse prognosis for patients with MBM was significantly associated with the number of metastases at diagnosis, previous exposure to immunotherapy, spread of disease to the leptomeningeal compartment, serum lactate dehydrogenase elevation, and the presence of extracranial disease. The current age of systemic treatments has also been accompanied by shifts in the use of central nervous system-directed therapies.
Collapse
Affiliation(s)
- Evan D Bander
- Department of Neurosurgery and Brain Metastasis Center, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Neurosurgery, New York Presbyterian Hospital/Weill Cornell Medical College, New York, New York
| | - Melissa Yuan
- Department of Neurosurgery and Brain Metastasis Center, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joseph A Carnevale
- Department of Neurosurgery and Brain Metastasis Center, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Neurosurgery, New York Presbyterian Hospital/Weill Cornell Medical College, New York, New York
| | - Anne S Reiner
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Katherine S Panageas
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael A Postow
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Viviane Tabar
- Department of Neurosurgery and Brain Metastasis Center, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nelson S Moss
- Department of Neurosurgery and Brain Metastasis Center, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
648
|
Martín-Soberón MC, Ruiz S, De Velasco G, Yarza R, Carretero A, Castellano D, Sepúlveda-Sánchez JM. Pneumatosis intestinalis in a radioactive iodine-refractory metastasic thyroid papillary carcinoma with BRAF V600E mutation treated with dabrafenib-trametinib: a case report. J Med Case Rep 2021; 15:109. [PMID: 33653337 PMCID: PMC7927265 DOI: 10.1186/s13256-020-02581-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/11/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Pneumatosis intestinalis (PI) is a rare entity which refers to the presence of gas within the wall of the small bowel or colon which is a radiographic sign. The etiology and clinical presentation are variable. Patients with PI may present either with chronic mild non-specific symptoms or with acute abdominal pain with peritonitis. Some cases of intestinal pneumatosis have been reported as adverse events of new oncological treatments such as targeted therapies that are widely used in multiple tumors. CASE PRESENTATION A 59-year-old caucasian female with radioactive iodine-refractory metastatic thyroid papillary carcinoma with BRAFV600E mutation was treated with dabrafenib and trametinib as a compassionate use. After 4 months treatment, positron emission tomography-computed tomography (PET-CT) showed PI. At the time of diagnosis, the patient was asymptomatic without signs of peritonitis. The initial treatment was conservative and no specific treatment for PI was needed. Unfortunately, after dabrafenib-trametinib withdrawal, the patient developed tumor progression with significant clinical worsening. CONCLUSIONS This case report is, in our knowledge, the first description of PI in a patient treated with dabrafenib-trametinib. Conservative treatment is feasible if there are no abdominal symptoms.
Collapse
Affiliation(s)
- M C Martín-Soberón
- Medical Oncology Department, University Hospital 12 de Octubre, Madrid, Spain.
| | - S Ruiz
- Nuclear Medicine Department, University Hospital 12 de Octubre, Madrid, Spain
| | - G De Velasco
- Medical Oncology Department, University Hospital 12 de Octubre, Madrid, Spain
| | - R Yarza
- Medical Oncology Department, University Hospital 12 de Octubre, Madrid, Spain
| | - A Carretero
- Medical Oncology Department, University Hospital 12 de Octubre, Madrid, Spain
| | - D Castellano
- Medical Oncology Department, University Hospital 12 de Octubre, Madrid, Spain
| | | |
Collapse
|
649
|
Padder RA, Bhat ZI, Ahmad Z, Singh N, Husain M. DRP1 Promotes BRAF V600E-Driven Tumor Progression and Metabolic Reprogramming in Colorectal Cancer. Front Oncol 2021; 10:592130. [PMID: 33738242 PMCID: PMC7961078 DOI: 10.3389/fonc.2020.592130] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 12/30/2020] [Indexed: 12/12/2022] Open
Abstract
Background Mitochondria are highly dynamic organelles which remain in a continuous state of fission/ fusion dynamics to meet the metabolic needs of a cell. However, this fission/fusion dynamism has been reported to be dysregulated in most cancers. Such enhanced mitochondrial fission is demonstrated to be positively regulated by some activating oncogenic mutations; such as those of KRAS (Kristen rat sarcoma viral oncogene homologue) or BRAF (B- rapidly accelerated fibrosarcoma), thereby increasing tumor progression/ chemotherapeutic resistance and metabolic deregulation. However, the underlying mechanism(s) are still not clear, thus highlighting the need to further explore possible mechanism(s) of intervention. We sought to investigate how BRAFV600E driven CRC (colorectal cancer) progression is linked to mitochondrial fission/fusion dynamics and whether this window could be exploited to target CRC progression. Methods Western blotting was employed to study the differences in expression levels of key proteins regulating mitochondrial dynamics, which was further confirmed by confocal microscopy imaging of mitochondria in endogenously expressing BRAFWT and BRAFV600E CRC cells. Proliferation assays, soft agar clonogenic assays, glucose uptake/lactate production, ATP/ NADPH measurement assays were employed to study the extent of carcinogenesis and metabolic reprograming in BRAFV600E CRC cells. Genetic knockdown (shRNA/ siRNA) and/or pharmacologic inhibition of Dynamin related protein1/Pyruvate dehydrogenase kinase1 (DRP1/PDK1) and/or BRAFV600E were employed to study the involvement and possible mechanism of these proteins in BRAFV600E driven CRC. Statistical analyses were carried out using Graph Pad Prism v 5.0, data was analyzed by unpaired t-test and two-way ANOVA with appropriate post hoc tests. Results Our results demonstrate that BRAFV600E CRC cells have higher protein levels of mitochondrial fission factor- DRP1/pDRP1S616 leading to a more fragmented mitochondrial state compared to those harboring BRAFWT . This fragmented mitochondrial state was found to confer glycolytic phenotype, clonogenic potential and metastatic advantage to cells harboring BRAFV600E . Interestingly, such fragmented mitochondrial state seemed positively regulated by mitochondrial PDK1 as observed through pharmacologic as well as genetic inhibition of PDK1. Conclusion In conclusion, our data suggest that BRAFV600E driven colorectal cancers have fragmented mitochondria which confers glycolytic phenotype and growth advantage to these tumors, and such phenotype is dependent at least in part on PDK1- thus highlighting a potential therapeutic target.
Collapse
Affiliation(s)
- Rayees Ahmad Padder
- 409-Cancer Biology Laboratory, Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Zafar Iqbal Bhat
- Department of Zoology, PMB Gujrati Science College, Devi Ahilya Vishwavidyalaya, Indore, India
| | - Zaki Ahmad
- 409-Cancer Biology Laboratory, Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Neetu Singh
- Advanced Instrumentation Research Facility, Jawaharlal Nehru University, New Delhi, India
| | - Mohammad Husain
- 409-Cancer Biology Laboratory, Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
650
|
Abstract
PURPOSE OF REVIEW BRAF/MEK inhibitor has changed the treatment landscape in patients with advanced and metastatic melanoma with prolonged overall survival and progression-free survival. Since three treatment combinations exist with similar efficacy therapy decisions are often made based on the side effect profile. Additionally, on-target side effects or class effects have to be properly managed to ensure treatment adherence. RECENT FINDINGS Sequential treatment with BRAF/MEK inhibition and immunotherapy might increase toxicity with a sepsis-like syndrome and triple therapy with concomitant BRAF/MEK inhibition and anti-PD1/PD-L1 antibody therapy induces severe side effects in the vast majority of patients. SUMMARY Toxicity of combination therapy with BRAF/MEK inhibitors is generally manageable, reversible and infrequently associated with treatment discontinuation. In case of persisting off-target effects the change to another combination therapy can resolve side effects.
Collapse
Affiliation(s)
- Alvaro Moreira
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai
- The Kimberly and Eric J. Waldman Department of Dermatology at Mount Sinai, New York, NY, USA
| | - Céleste Lebbé
- Université de Paris, AP-HP Dermatology, INSERM U976, Saint Louis Hospital, Paris, France
| | - Lucie Heinzerling
- Department of Dermatology, Universitätsklinikum München (LMU), Munich, Germany
- Department of Dermatology, Universitätsklinikum Erlangen, Germany and Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Erlangen, Germany
| |
Collapse
|