1
|
Wang S, Riedstra CP, Zhang Y, Anandh S, Dudley AC. PTEN-restoration abrogates brain colonisation and perivascular niche invasion by melanoma cells. Br J Cancer 2024; 130:555-567. [PMID: 38148377 PMCID: PMC10876963 DOI: 10.1038/s41416-023-02530-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/28/2023] Open
Abstract
BACKGROUND Melanoma brain metastases (MBM) continue to be a significant clinical problem with limited treatment options. Highly invasive melanoma cells migrate along the vasculature and perivascular cells may contribute to residual disease and recurrence. PTEN loss and hyperactivation of AKT occur in MBM; however, a role for PTEN/AKT in perivascular invasion has not been described. METHODS We used in vivo intracranial injections of murine melanoma and bulk RNA sequencing of melanoma cells co-cultured with brain endothelial cells (brECs) to investigate brain colonisation and perivascular invasion. RESULTS We found that PTEN-null melanoma cells were highly efficient at colonising the perivascular niche relative to PTEN-expressing counterparts. PTEN re-expression (PTEN-RE) in melanoma cells significantly reduced brain colonisation and migration along the vasculature. We hypothesised this phenotype was mediated through vascular-induced TGFβ secretion, which drives AKT phosphorylation. Disabling TGFβ signalling in melanoma cells reduced colonisation and perivascular invasion; however, the introduction of constitutively active myristolated-AKT (myrAKT) restored overall tumour size but not perivascular invasion. CONCLUSIONS PTEN loss facilitates perivascular brain colonisation and invasion of melanoma. TGFβ-AKT signalling partially contributes to this phenotype, but further studies are needed to determine the complementary mechanisms that enable melanoma cells to both survive and spread along the brain vasculature.
Collapse
Affiliation(s)
- Sarah Wang
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia, Charlottesville, VA, 22908, USA
| | - Caroline P Riedstra
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia, Charlottesville, VA, 22908, USA
| | - Yu Zhang
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia, Charlottesville, VA, 22908, USA
| | - Swetha Anandh
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia, Charlottesville, VA, 22908, USA
| | - Andrew C Dudley
- Department of Microbiology, Immunology, and Cancer Biology, The University of Virginia, Charlottesville, VA, 22908, USA.
- The University of Virginia Comprehensive Cancer Center, Charlottesville, VA, USA.
| |
Collapse
|
2
|
Bachari A, Nassar N, Schanknecht E, Telukutla S, Piva TJ, Mantri N. Rationalizing a prospective coupling effect of cannabinoids with the current pharmacotherapy for melanoma treatment. WIREs Mech Dis 2024; 16:e1633. [PMID: 37920964 DOI: 10.1002/wsbm.1633] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/21/2023] [Accepted: 10/06/2023] [Indexed: 11/04/2023]
Abstract
Melanoma is one of the leading fatal forms of cancer, yet from a treatment perspective, we have minimal control over its reoccurrence and resistance to current pharmacotherapies. The endocannabinoid system (ECS) has recently been accepted as a multifaceted homeostatic regulator, influencing various physiological processes across different biological compartments, including the skin. This review presents an overview of the pathophysiology of melanoma, current pharmacotherapy used for treatment, and the challenges associated with the different pharmacological approaches. Furthermore, it highlights the utility of cannabinoids as an additive remedy for melanoma by restoring the balance between downregulated immunomodulatory pathways and elevated inflammatory cytokines during chronic skin conditions as one of the suggested critical approaches in treating this immunogenic tumor. This article is categorized under: Cancer > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Ava Bachari
- The Pangenomics Lab, School of Science, RMIT University, Bundoora, Victoria, Australia
| | - Nazim Nassar
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Ellen Schanknecht
- The Pangenomics Lab, School of Science, RMIT University, Bundoora, Victoria, Australia
| | | | - Terrence Jerald Piva
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Nitin Mantri
- The Pangenomics Lab, School of Science, RMIT University, Bundoora, Victoria, Australia
- The UWA Institute of Agriculture, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
3
|
Yu Q, Ma Y, Feng T. A 41-Year-Old Woman with a Late Cerebral Metastasis 16 Years After an Initial Diagnosis of Cutaneous Melanoma. Am J Case Rep 2022; 23:e935728. [PMID: 35256582 PMCID: PMC8919240 DOI: 10.12659/ajcr.935728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Patient: Female, 41-year-old
Final Diagnosis: Melanoma brain metastasis
Symptoms: Intrercranial hemorraghe
Medication:—
Clinical Procedure: —
Specialty: Neurosurgery
Collapse
Affiliation(s)
- Qi Yu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Yawen Ma
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Tianda Feng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| |
Collapse
|
4
|
Tawbi HA, Forsyth PA, Hodi FS, Algazi AP, Hamid O, Lao CD, Moschos SJ, Atkins MB, Lewis K, Postow MA, Thomas RP, Glaspy J, Jang S, Khushalani NI, Pavlick AC, Ernstoff MS, Reardon DA, Kudchadkar R, Tarhini A, Chung C, Ritchings C, Durani P, Askelson M, Puzanov I, Margolin KA. Long-term outcomes of patients with active melanoma brain metastases treated with combination nivolumab plus ipilimumab (CheckMate 204): final results of an open-label, multicentre, phase 2 study. Lancet Oncol 2021; 22:1692-1704. [PMID: 34774225 DOI: 10.1016/s1470-2045(21)00545-3] [Citation(s) in RCA: 184] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Combination nivolumab plus ipilimumab was efficacious in patients with asymptomatic melanoma brain metastases (MBM) in CheckMate 204, but showed low efficacy in patients with symptomatic MBM. Here, we provide final 3-year follow-up data from the trial. METHODS This open-label, multicentre, phase 2 study (CheckMate 204) included adults (aged ≥18 years) with measurable MBM (0·5-3·0 cm in diameter). Asymptomatic patients (cohort A) had an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1 and no neurological symptoms or baseline corticosteroid use; symptomatic patients (cohort B) had an ECOG performance status of 0-2 with stable neurological symptoms and could be receiving low-dose dexamethasone. Nivolumab 1 mg/kg plus ipilimumab 3 mg/kg was given intravenously every 3 weeks for four doses, followed by nivolumab 3 mg/kg every 2 weeks for up to 2 years, until disease progression or unacceptable toxicity. The primary endpoint was intracranial clinical benefit rate (complete responses, partial responses, or stable disease lasting ≥6 months) assessed in all treated patients. Intracranial progression-free survival and overall survival were key secondary endpoints. This study is registered with ClinicalTrials.gov, NCT02320058. FINDINGS Between Feb 19, 2015, and Nov 1, 2017, 119 (72%) of 165 screened patients were enrolled and treated: 101 patients were asymptomatic (cohort A; median follow-up 34·3 months [IQR 14·7-36·4]) and 18 were symptomatic (cohort B; median follow-up 7·5 months [1·2-35·2]). Investigator-assessed intracranial clinical benefit was observed in 58 (57·4% [95% CI 47·2-67·2]) of 101 patients in cohort A and three (16·7% [3·6-41·4]) of 18 patients in cohort B; investigator-assessed objective response was observed in 54 (53·5% [43·3-63·5]) patients in cohort A and three (16·7% [3·6-41·4]) patients in cohort B. 33 (33%) patients in cohort A and three (17%) patients in cohort B had an investigator-assessed intracranial complete response. For patients in cohort A, 36-month intracranial progression-free survival was 54·1% (95% CI 42·7-64·1) and overall survival was 71·9% (61·8-79·8). For patients in cohort B, 36-month intracranial progression-free survival was 18·9% (95% CI 4·6-40·5) and overall survival was 36·6% (14·0-59·8). The most common grade 3-4 treatment-related adverse events (TRAEs) were increased alanine aminotransferase and aspartate aminotransferase (15 [15%] of 101 patients each) in cohort A; no grade 3 TRAEs occurred in more than one patient each in cohort B, and no grade 4 events occurred. The most common serious TRAEs were colitis, diarrhoea, hypophysitis, and increased alanine aminotransferase (five [5%] of each among the 101 patients in cohort A); no serious TRAE occurred in more than one patient each in cohort B. There was one treatment-related death (myocarditis in cohort A). INTERPRETATION The durable 3-year response, overall survival, and progression-free survival rates for asymptomatic patients support first-line use of nivolumab plus ipilimumab. Symptomatic disease in patients with MBM remains difficult to treat, but some patients achieve a long-term response with the combination. FUNDING Bristol Myers Squibb.
Collapse
Affiliation(s)
- Hussein A Tawbi
- University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Peter A Forsyth
- Department of Neuro-Oncology, Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | | | - Alain P Algazi
- Melanoma Center, University of California-San Francisco, San Francisco, CA, USA
| | - Omid Hamid
- Melanoma Center, The Angeles Clinic and Research Institute, Los Angeles, CA, USA
| | - Christopher D Lao
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Stergios J Moschos
- Division of Hematology & Oncology, The University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | - Michael B Atkins
- Department of Medical Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Washington DC, USA
| | - Karl Lewis
- Department of Medical Oncology, University of Colorado Comprehensive Cancer Center, Aurora, CO, USA
| | - Michael A Postow
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Reena P Thomas
- Department of Neurology, Stanford University Cancer Center, Stanford, CA, USA
| | - John Glaspy
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
| | - Sekwon Jang
- Inova Schar Cancer Institute, Fairfax, VA, USA
| | - Nikhil I Khushalani
- Department of Cutaneous Oncology, H Lee Moffitt Cancer Center, Tampa, FL USA
| | - Anna C Pavlick
- Department of Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Marc S Ernstoff
- Department of Immuno-Oncology, Division of Cancer Treatment and Diagnosis, National Cancer Institute at the National Institutes of Health, Rockville, MD, USA
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ragini Kudchadkar
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Ahmad Tarhini
- Departments of Cutaneous Oncology and Immunology, Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Caroline Chung
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | - Igor Puzanov
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kim A Margolin
- Department of Medical Oncology, City of Hope, Duarte, CA, USA
| |
Collapse
|
5
|
Steindl A, Brastianos PK, Preusser M, Berghoff AS. Precision medicine biomarkers in brain metastases: applications, discordances, and obstacles. Neurooncol Adv 2021; 3:v35-v42. [PMID: 34859231 PMCID: PMC8633753 DOI: 10.1093/noajnl/vdab105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Brain metastases (BM) present a common cause of mortality and morbidity in several metastatic cancer entities. New therapeutic developments during the last decades, including targeted and immune-related therapies, have shown considerable extra- and intracranial response rates in specific subgroups of BM patients. However, differences in the molecular alteration in the BM tumor tissue compared to extracranial tumors leads to heterogeneous therapeutic responses. Therefore, an accurate molecular analyzation of BM tissue, if possible, has become an essential part in therapeutic decision making in BM patients. The concordance of predictive molecular biomarkers between multiple sites including extracranial and intracranial tumor tissue have been analyzed for some but not all biomarkers routinely applied in modern precision medicine approaches. In the present review, we summarize the current evidence of predictive biomarkers for personalized therapy approaches in the treatment of parenchymal BM.
Collapse
Affiliation(s)
- Ariane Steindl
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Priscilla K Brastianos
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Division of Neuro-Oncology, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Anna S Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
6
|
Steindl A, Berghoff AS. Brain metastases in metastatic cancer: a review of recent advances in systemic therapies. Expert Rev Anticancer Ther 2020; 21:325-339. [PMID: 33196341 DOI: 10.1080/14737140.2021.1851200] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction: Brain metastases (BM) are a frequent complication of metastatic cancer. Due to the wider availability and application of screening procedures, an increasing fraction of patients are diagnosed at the asymptomatic stage. The introduction of immune checkpoint inhibitors and targeted therapies has revolutionized treatment in several frequently BM-causing entities like metastatic lung cancer, melanoma and breast cancer. However, registered trials of new targeted and immunotherapy mostly excluded patients with BM resulting in limited knowledge of the intracranial efficacy of new systemic agents.Areas covered: The present review highlights recent advances in systemic therapies for the treatment and prophylaxis of the three leading BM causing tumors: NSCLC, melanoma and breast cancer.Expert opinion: High intracranial efficacy was observed for several next-generation tyrosine kinase inhibitors as well as immune checkpoint inhibitors, especially in patients with asymptomatic disease. Ongoing discussions addressed the need for local therapies in patients with asymptomatic BM and the availability of systemic therapy with high intracranial efficacy. Further BM-specific studies as well as BM-specific endpoints in registered trials are needed to define the role of systemic monotherapies in patients with BM.
Collapse
Affiliation(s)
- Ariane Steindl
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Anna S Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
7
|
Abstract
The development of brain metastases occurs in 10–20% of all patients with cancer. Brain metastases portend poor survival and contribute to increased cancer mortality and morbidity. Despite multimodal treatment options, which include surgery, radiotherapy, and chemotherapy, 5-year survival remains low. Besides, our current treatment modalities can have significant neurological comorbidities, which result in neurocognitive decline and a decrease in a patient’s quality of life. However, innovations in technology, improved understanding of tumor biology, and new therapeutic options have led to improved patient care. Novel approaches in radiotherapy are minimizing the neurocognitive decline while providing the same therapeutic benefit. In addition, advances in targeted therapies and immune checkpoint inhibitors are redefining the management of lung and melanoma brain metastases. Similar approaches to brain metastases from other primary tumors promise to lead to new and effective therapies. We are beginning to understand the appropriate combination of these novel approaches with our traditional treatment options. As advances in basic and translational science and innovative technologies enter clinical practice, the prognosis of patients with brain metastases will continue to improve.
Collapse
Affiliation(s)
- Adam Lauko
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
| | - Yasmeen Rauf
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
| | - Manmeet S Ahluwalia
- Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
8
|
Simonsen TG, Gaustad JV, Rofstad EK. Bevacizumab treatment of meningeal melanoma metastases. J Transl Med 2020; 18:13. [PMID: 31915016 PMCID: PMC6947957 DOI: 10.1186/s12967-020-02212-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/03/2020] [Indexed: 12/31/2022] Open
Abstract
Background Melanoma patients with metastatic growth in the meninges have poor prognosis and few treatment options. Although treatment with BRAF inhibitors or immune checkpoint inhibitors has provided promising results, most patients with advanced melanoma are resistant to these treatments and develop severe side effects. Novel treatment strategies are needed for patients with meningeal melanoma metastases, and the potential of antiangiogenic therapy was investigated in this preclinical study. Methods Two GFP-transfected melanoma models (A-07 and D-12) differing substantially in VEGF-A expression were included in the study, and the anti-VEGF-A antibody bevacizumab was used as therapeutic agent. Meningeal metastases were initiated in BALB/c nu/nu mice by intracranial inoculation of melanoma cells, and bevacizumab treatment was given twice a week in i.p. doses of 10 mg/kg until the mice became moribund. Therapeutic effects were evaluated by determining tumor host survival time, assessing tumor growth and angiogenic activity by quantitative analyses of histological preparations, and measuring the expression of angiogenesis-related genes by quantitative PCR. Results Meningeal A-07 melanomas showed higher expression of VEGF-A than meningeal D-12 melanomas, whereas the expression of ANGPT2 and IL8, two important angiogenesis drivers in melanoma, was much higher in D-12 than in A-07 tumors. Bevacizumab treatment inhibited tumor angiogenesis and prolonged host survival in mice with A-07 tumors but not in mice with D-12 tumors. Meningeal A-07 tumors in bevacizumab-treated mice compensated for the reduced VEGF-A activity by up-regulating a large number of angiogenesis-related genes, including ANGPT2 and its receptors TIE1 and TIE2. Melanoma cells migrated from meningeal tumors into the cerebrum, where they initiated metastatic growth by vessel co-option. In the A-07 model, the density of cerebral micrometastases was higher in bevacizumab-treated than in untreated mice, either because bevacizumab treatment increased mouse survival or induced increased tumor gene expression. Conclusions The development of antiangiogenic strategies for the treatment of meningeal melanoma metastases is a challenging task because the outcome of treatment will depend on the angiogenic signature of the tumor tissue, treatment-induced alterations of the angiogenic signature, and the treatment sensitivity of metastatic lesions in other intracranial sites.
Collapse
Affiliation(s)
- Trude G Simonsen
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Jon-Vidar Gaustad
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Einar K Rofstad
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
9
|
Delou JMA, Souza ASO, Souza LCM, Borges HL. Highlights in Resistance Mechanism Pathways for Combination Therapy. Cells 2019; 8:E1013. [PMID: 31480389 PMCID: PMC6770082 DOI: 10.3390/cells8091013] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 12/14/2022] Open
Abstract
Combination chemotherapy has been a mainstay in cancer treatment for the last 60 years. Although the mechanisms of action and signaling pathways affected by most treatments with single antineoplastic agents might be relatively well understood, most combinations remain poorly understood. This review presents the most common alterations of signaling pathways in response to cytotoxic and targeted anticancer drug treatments, with a discussion of how the knowledge of signaling pathways might support and orient the development of innovative strategies for anticancer combination therapy. The ultimate goal is to highlight possible strategies of chemotherapy combinations based on the signaling pathways associated with the resistance mechanisms against anticancer drugs to maximize the selective induction of cancer cell death. We consider this review an extensive compilation of updated known information on chemotherapy resistance mechanisms to promote new combination therapies to be to discussed and tested.
Collapse
Affiliation(s)
- João M A Delou
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Alana S O Souza
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Leonel C M Souza
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Helena L Borges
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.
| |
Collapse
|
10
|
Chukwueke UN, Brastianos PK. Precision Medical Approaches to the Diagnoses and Management of Brain Metastases. Curr Treat Options Oncol 2019; 20:49. [PMID: 31062107 DOI: 10.1007/s11864-019-0649-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OPINION STATEMENT Brain metastases represent a common and devastating complication of cancer with survival on the order of a few months in most patients. Melanoma, breast cancer, and lung cancer remain the primary disease histologies with the highest rates of metastatic spread to the brain. The incidence of brain metastases has continued to rise, likely explained by multiple factors. Improvement in progression-free survival in systemic cancer is likely attributable to advances in medical therapy, earlier supportive and symptomatic care, and improved precision around diagnosis and detection. In this context, longer survival and improved extracranial control disease has likely contributed to the increased development of metastatic spread intracranially. The foundation of management remains systemic therapy, as well as a combination of surgery and radiation therapy. In the era of targeted therapies, specific agents have demonstrated improved CNS penetration, however with varying degrees of durable responses. Most patients develop resistance to targeted agents, limiting their duration of use for patients. In this era of personalized medicine, the role of genomic characterization in cancer has been critical in the field of brain metastases, as alterations unique to both the brain metastases and its systemic predecessor have been identified, potentially offering new avenues for therapy.
Collapse
Affiliation(s)
- Ugonma N Chukwueke
- Center for Neuro-Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA. .,Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215, USA.
| | - Priscilla K Brastianos
- Department of Medical Oncology, Division of Neuro-Oncology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA.,Department of Medicine, Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| |
Collapse
|
11
|
Schwarz D, Niederle T, Münch P, Hielscher T, Hassel JC, Schlemmer HP, Platten M, Winkler F, Wick W, Heiland S, Delorme S, Bendszus M, Bäumer P, Breckwoldt MO. Susceptibility-weighted imaging in malignant melanoma brain metastasis. J Magn Reson Imaging 2019; 50:1251-1259. [PMID: 30793419 DOI: 10.1002/jmri.26692] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 02/07/2019] [Accepted: 02/07/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The value of cerebral susceptibility-weighted imaging (SWI) in malignant melanoma (MM) patients remains controversial and the effect of melanin on SWI is not well understood. PURPOSE To systematically analyze the spectrum of intracerebral findings in MM brain metastases (BM) on SWI and to determine the diagnostic value of SWI. STUDY TYPE Retrospective. POPULATION/SUBJECTS In all, 100 patients with melanoma BM (69 having received radiotherapy [RT] and 31 RT-naïve) and a control group of 100 melanoma patients without BM were included. For detailed analysis of signal characteristics, 175 metastases were studied. FIELD STRENGTH/SEQUENCE Gradient echo SWI sequence at 1.5, 3.0, and 9.4 T. ASSESSMENT Signal characteristics from melanotic and amelanotic BMs on SWI with a focus on blooming artifacts were analyzed, as well as the presence and longitudinal dynamics of isolated SWI blooming artifacts in patients with and without BM. STATISTICAL TESTS Chi-squared and Student's t-test were used for contingency table measures and group data of signal and clinical characteristics, respectively. RESULTS Melanotic and amelanotic metastases did not show significant differences of SWI blooming artifacts (38% vs. 43%, P = 0.61). Most metastases without an initial SWI artifact developed a signal dropout during follow-up (80%; 65/81). Isolated SWI artifacts were detected more frequently in patients with BM (20 vs. 9, P = 0.03), of which the majority were found in patients who had received RT (17 vs. 3, P = 0.08). None of these isolated SWI blooming artifacts turned into overt metastases over time (median follow-up: 8.5 months). Similar findings persisted as remnants of successfully treated metastases (88%; 7/8). DATA CONCLUSION We conclude that SWI provides little additional diagnostic benefit over standard T1 -weighted imaging, as melanin content alone does not cause diagnostically relevant SWI blooming. Signal transition of SWI may rather indicate secondary phenomena like microbleeding and/or metal scavenging. Our results suggest that isolated SWI artifacts do not constitute vital tumor tissue but represent unspecific microbleedings, RT-related parenchymal changes or posttherapeutic remnants of former metastatic lesions. LEVEL OF EVIDENCE 3 Technical Efficacy Stage: 5 J. Magn. Reson. Imaging 2019;50:1251-1259.
Collapse
Affiliation(s)
- Daniel Schwarz
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany.,Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Niederle
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Philipp Münch
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Thomas Hielscher
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jessica C Hassel
- Dematology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Michael Platten
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany.,Neurology Clinic, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Frank Winkler
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Neurooncology, German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany
| | - Wolfgang Wick
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Neurooncology, German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany
| | - Sabine Heiland
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Delorme
- Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Philipp Bäumer
- Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael O Breckwoldt
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
12
|
Seoane J, De Mattos-Arruda L, Le Rhun E, Bardelli A, Weller M. Cerebrospinal fluid cell-free tumour DNA as a liquid biopsy for primary brain tumours and central nervous system metastases. Ann Oncol 2019; 30:211-218. [PMID: 30576421 DOI: 10.1093/annonc/mdy544] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Challenges in obtaining tissue specimens from patients with brain tumours limit the diagnosis and molecular characterisation and impair the development of better therapeutic approaches. The analysis of cell-free tumour DNA in plasma (considered a liquid biopsy) has facilitated the characterisation of extra-cranial tumours. However, cell-free tumour DNA in plasma is limited in quantity and may not reliably capture the landscape of genomic alterations of brain tumours. Here, we review recent work assessing the relevance of cell-free tumour DNA from cerebrospinal fluid in the characterisation of brain cancer. We focus on the advances in the use of the cerebrospinal fluid as a source of cell-free tumour DNA to facilitate diagnosis, reveal actionable genomic alterations, monitor responses to therapy, and capture tumour heterogeneity in patients with primary brain tumours and brain and leptomeningeal metastases. Profiling cerebrospinal fluid cell-free tumour DNA provides the opportunity to precisely acquire and monitor genomic information in real time and guide precision therapies.
Collapse
Affiliation(s)
- J Seoane
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona; CIBERONC, Barcelona; Universitat Autònoma de Barcelona, Cerdanyola del Vallès.
| | - L De Mattos-Arruda
- Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Barcelona
| | - E Le Rhun
- Lille University, Inserm U1192 PRISM, Villeneuve d'Ascq; Neuro-oncology, Department of Neurosurgery, University Hospital, Lille; Neuro-oncology, Breast Unit, Department of Medical Oncology, Oscar Lambret Center, Lille, France
| | - A Bardelli
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo (TO); Department of Oncology, University of Torino, Candiolo (TO), Italy
| | - M Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Chen X, Wang R, Chen A, Wang Y, Wang Y, Zhou J, Cao R. Inhibition of mouse RM-1 prostate cancer and B16F10 melanoma by the fusion protein of HSP65 & STEAP1 186-193. Biomed Pharmacother 2019; 111:1124-1131. [PMID: 30841425 DOI: 10.1016/j.biopha.2019.01.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/20/2018] [Accepted: 01/05/2019] [Indexed: 01/28/2023] Open
Abstract
The research of tumor vaccine plays a crucial role in tumor immunotherapy. This study has constructed and prepared a fusion protein vaccine of heat shock protein 65 (HSP65) and the octapeptide epitope 186-193 of the six transmembrane epithelial antigen of the prostate 1 (STEAP1 186-193), and investigated the inhibitory effect of the fusion protein on mouse RM-1 prostate cancer and B16F10 melanoma xenografts. The fusion protein His-HSP65-STEAP1 186-193 (HHST1), His-HSP65-2×STEAP1 186-193 (HHST2) and His-HSP65-6×STEAP1 186-193 (HHST6) were obtained by setting different copy number of STEAP1 186-193 and adding His purification tag before HSP65. Firstly the inhibitory effect of fusion protein on mouse RM-1 prostate cancer xenografts has been studied, which could be the basis of the study the inhibitory effect of the best fusion protein on mouse B16F10 melanoma xenografts. All studies compared with the fusion protein His-HSP65 (HHSP65), the fusion proteins HHST1, HHST2 and HHST6 all could significantly inhibit the growth of mouse RM-1 prostate cancer xenografts. In addition, the fusion protein HHST2 was proved to be the best compared with the fusion proteins HHST1 and HHST6 (P<0.05). Apart from this, compared with the fusion protein HHSP65, the fusion protein HHST2 also significantly inhibited the growth of mouse beared B16F10 melanoma. The results above indicate that HSP65 and STEAP1 186-193 can significantly inhibit the growth of mouse RM-1 prostate cancer and B16F10 melanoma xenografts, and the appropriate increase of copy number can effectively improve that the fusion protein has an excellent anti-tumor ability.
Collapse
Affiliation(s)
- Xuan Chen
- Microgene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, No. 24, Tongjia Alley, Central Road, Nanjing, 210009, China
| | - Rui Wang
- Microgene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, No. 24, Tongjia Alley, Central Road, Nanjing, 210009, China
| | - Anji Chen
- Microgene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, No. 24, Tongjia Alley, Central Road, Nanjing, 210009, China
| | - Yongmei Wang
- Microgene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, No. 24, Tongjia Alley, Central Road, Nanjing, 210009, China
| | - Yiqin Wang
- Microgene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, No. 24, Tongjia Alley, Central Road, Nanjing, 210009, China
| | - Jialei Zhou
- Microgene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, No. 24, Tongjia Alley, Central Road, Nanjing, 210009, China
| | - Rongyue Cao
- Microgene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, No. 24, Tongjia Alley, Central Road, Nanjing, 210009, China.
| |
Collapse
|
14
|
Brastianos PK, Ippen FM, Hafeez U, Gan HK. Emerging Gene Fusion Drivers in Primary and Metastatic Central Nervous System Malignancies: A Review of Available Evidence for Systemic Targeted Therapies. Oncologist 2018; 23:1063-1075. [PMID: 29703764 PMCID: PMC6192601 DOI: 10.1634/theoncologist.2017-0614] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/07/2018] [Indexed: 12/11/2022] Open
Abstract
Primary and metastatic tumors of the central nervous system present a difficult clinical challenge, and they are a common cause of disease progression and death. For most patients, treatment consists primarily of surgery and/or radiotherapy. In recent years, systemic therapies have become available or are under investigation for patients whose tumors are driven by specific genetic alterations, and some of these targeted treatments have been associated with dramatic improvements in extracranial and intracranial disease control and survival. However, the success of other systemic therapies has been hindered by inadequate penetration of the drug into the brain parenchyma. Advances in molecular characterization of oncogenic drivers have led to the identification of new gene fusions driving oncogenesis in some of the most common sources of intracranial tumors. Systemic therapies targeting many of these alterations have been approved recently or are in clinical development, and the ability to penetrate the blood-brain barrier is now widely recognized as an important property of such drugs. We review this rapidly advancing field with a focus on recently uncovered gene fusions and brain-penetrant systemic therapies targeting them. IMPLICATIONS FOR PRACTICE Driver gene fusions involving receptor tyrosine kinases have been identified across a wide range of tumor types, including primary central nervous system (CNS) tumors and extracranial solid tumors that are associated with high rates of metastasis to the CNS (e.g., lung, breast, melanoma). This review discusses the systemic therapies that target emerging gene fusions, with a focus on brain-penetrant agents that will target the intracranial disease and, where present, also extracranial disease.
Collapse
Affiliation(s)
- Priscilla K Brastianos
- Department of Hematology and Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Franziska Maria Ippen
- Department of Hematology and Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Umbreen Hafeez
- Medical Oncology, Austin Hospital, Heidelberg, Melbourne, Australia
| | - Hui K Gan
- Medical Oncology, Austin Hospital, Heidelberg, Melbourne, Australia
- La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia
- Department of Medicine, University of Melbourne, Heidelberg, Victoria, Australia
| |
Collapse
|
15
|
Xia Y, Mashouf LA, Maxwell R, Peng LC, Lipson EJ, Sharfman WH, Bettegowda C, Redmond KJ, Kleinberg LR, Lim M. Adjuvant radiotherapy and outcomes of presumed hemorrhagic melanoma brain metastases without malignant cells. Surg Neurol Int 2018; 9:146. [PMID: 30105140 PMCID: PMC6080145 DOI: 10.4103/sni.sni_140_18] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 06/14/2018] [Indexed: 12/28/2022] Open
Abstract
Background Patients with melanoma can present with a hemorrhagic intracranial lesion. Upon resection, pathology reports may not detect any malignant cells. However, the hemorrhage may obscure their presence and so physicians may still decide whether adjuvant radiotherapy should be applied. Here, we report on the outcomes of a series of patients with melanoma with hemorrhagic brain lesions that returned with no tumor cells. Methods All melanoma patients who had craniotomies from 2008 to 2017 at a single institution for hemorrhagic brain lesions were identified through retrospective chart review. Those who had pathology reports with no malignant cells were analyzed. Recurrence at the former site of hemorrhage and resection was the primary outcome. Results Ten patients met inclusion criteria, and the median follow-up time was 8.5 (1.8-27.3) months. At the time of craniotomy, the median number of brain lesions was 3 (1-25). Two patients had prior craniotomies, eight had prior radiation, and six had prior immunotherapy to the lesion of interest. After surgery, one patient received stereotactic radiosurgery (SRS) to the resection bed. Only one patient developed subsequent melanoma at the resection site; this patient developed the lesion recurrence once and had not received postoperative SRS. Conclusion Although small foci of metastatic disease as a source of bleeding for some patients cannot be excluded, melanoma patients with a suspected hemorrhagic brain metastasis that shows no tumor cells on pathology may benefit from close observation. The local recurrence risk in such cases appears to be low, even without adjuvant radiation.
Collapse
Affiliation(s)
- Yuanxuan Xia
- Department of Neurosurgery, Johns Hopkins Medical Institutes, Baltimore, Maryland, USA
| | - Leila A Mashouf
- Department of Neurosurgery, Johns Hopkins Medical Institutes, Baltimore, Maryland, USA
| | - Russell Maxwell
- Department of Neurosurgery, Johns Hopkins Medical Institutes, Baltimore, Maryland, USA.,Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Medical Institutes, Baltimore, Maryland, USA
| | - Luke C Peng
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Medical Institutes, Baltimore, Maryland, USA
| | - Evan J Lipson
- Department of Oncology, Johns Hopkins Medical Institutes, Baltimore, Maryland, USA
| | - William H Sharfman
- Department of Oncology, Johns Hopkins Medical Institutes, Baltimore, Maryland, USA
| | - Chetan Bettegowda
- Department of Neurosurgery, Johns Hopkins Medical Institutes, Baltimore, Maryland, USA
| | - Kristin J Redmond
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Medical Institutes, Baltimore, Maryland, USA
| | - Lawrence R Kleinberg
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Medical Institutes, Baltimore, Maryland, USA
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins Medical Institutes, Baltimore, Maryland, USA.,Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Medical Institutes, Baltimore, Maryland, USA.,Department of Oncology, Johns Hopkins Medical Institutes, Baltimore, Maryland, USA
| |
Collapse
|
16
|
Pruitt AA. Epidemiology, Treatment, and Complications of Central Nervous System Metastases. Continuum (Minneap Minn) 2018; 23:1580-1600. [PMID: 29200112 DOI: 10.1212/con.0000000000000551] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Neurologic problems resulting from systemic cancer metastases to brain parenchyma, dura, spinal cord, and leptomeninges are among the most common types of consultations addressed by neurologists. With patients surviving longer from systemic cancer, along with the rapidly evolving therapeutic options, the treatment of these devastating complications has become both more effective and more complicated. This article reviews current patterns of metastatic disease and the increasingly nuanced landscape of evolving therapies, their complications, and their impact on quality of survival. RECENT FINDINGS Targeted therapies with tyrosine kinase inhibitors and immune checkpoint inhibitors and cytotoxic therapies directed at disease-specific chemosensitivity patterns have dramatically improved the prognosis of non-small cell lung cancer, melanoma, and breast cancer, but have led to some novel complications and altered recurrence patterns. Clinical trials suggest the superiority of hippocampal-avoidance radiation fields and the use of stereotactic radiosurgery over whole-brain radiation therapy to minimize long-term cognitive consequences of radiation therapy. Emerging data document tolerable safety when brain radiation is combined with immunotherapy. Chemotherapy can be a first-line treatment for some inoperable brain metastases, eliminating or deferring whole-brain radiation therapy. Stereotactic body radiation therapy is a new technique of radiation used for spinal and epidural metastases that spares spinal cord tissue while ablating tumors. SUMMARY Metastases to the nervous system remain devastating, but their prognosis and therapies are more heterogeneous than previously appreciated. Neurologists now can offer more personalized prognostic information based on new stratification criteria, can predict drug complications relevant to the nervous system, and can provide critical partnership in the multidisciplinary effort to balance effective longer-term disease control with treatment-related adverse consequences.
Collapse
|
17
|
Furtner J, Berghoff AS, Schöpf V, Reumann R, Pascher B, Woitek R, Asenbaum U, Pelster S, Leitner J, Widhalm G, Gatterbauer B, Dieckmann K, Höller C, Prayer D, Preusser M. Temporal muscle thickness is an independent prognostic marker in melanoma patients with newly diagnosed brain metastases. J Neurooncol 2018; 140:173-178. [PMID: 30008154 PMCID: PMC6182383 DOI: 10.1007/s11060-018-2948-8] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 07/09/2018] [Indexed: 01/06/2023]
Abstract
Objectives The purpose of this study was to evaluate the prognostic relevance of temporal muscle thickness (TMT) in melanoma patients with newly diagnosed brain metastases. Methods TMT was retrospectively assessed in 146 melanoma patients with newly diagnosed brain metastases on cranial magnetic resonance images. Chart review was used to retrieve clinical parameters, including disease-specific graded prognostic assessment (DS-GPA) and survival times. Results Patients with a TMT > median showed a statistically significant increase in survival time (13 months) compared to patients with a TMT < median (5 months; p < 0.001; log rank test). A Cox regression model revealed that the risk of death was increased by 27.9% with every millimeter reduction in TMT. In the multivariate analysis, TMT (HR 0.724; 95% 0.642–0.816; < 0.001) and DS-GPA (HR 1.214; 95% CI 1.023–1.439; p = 0.026) showed a statistically significant correlation with overall survival. Conclusion TMT is an independent predictor of survival in melanoma patients with brain metastases. This parameter may aid in patient selection for clinical trials or to the choice of different treatment options based on the determination of frail patient populations.
Collapse
Affiliation(s)
- Julia Furtner
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Anna S Berghoff
- Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Veronika Schöpf
- Institute of Psychology, University of Graz, Universitaetsplatz 2, 8010, Graz, Austria.,BioTechMed, Mozartgasse 12, 8010, Graz, Austria
| | - Robert Reumann
- Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Benjamin Pascher
- Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Ramona Woitek
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Ulrika Asenbaum
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Sebastian Pelster
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Johannes Leitner
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Georg Widhalm
- Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Department of Neurosurgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Brigitte Gatterbauer
- Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Department of Neurosurgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Karin Dieckmann
- Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Department of Radiotherapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Christoph Höller
- Department of Dermatology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Daniela Prayer
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Matthias Preusser
- Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria. .,Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
18
|
Berghoff AS, Preusser M. New developments in brain metastases. Ther Adv Neurol Disord 2018; 11:1756286418785502. [PMID: 30034538 PMCID: PMC6048670 DOI: 10.1177/1756286418785502] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 04/11/2018] [Indexed: 12/25/2022] Open
Abstract
Patients with brain metastases (BM) are a population of high clinical need for new therapeutic approaches due to, as yet, very impaired survival prognosis. However, only few clinical trials have specifically addressed this prognostically highly heterogeneous patient population. New developments in the treatment of BM patients aim to reduce the side effects of local therapies, for example, by redefining the indications for stereotactic radiosurgery and whole-brain radiotherapy (WBRT) or introducing new applications like hippocampal sparing WBRT. Furthermore, systemic therapies become a more important treatment approach in patients harboring targetable mutations, as recent BM-specific endpoints in several phase III trials have shown promising intracranial efficacy. In addition, immune-checkpoint inhibitors show promising intracranial efficacy, particularly in patients with melanoma and non-small lung cancer BM. Here, we provide a review on the recent new developments in the local and systemic therapy approaches in BM patients.
Collapse
Affiliation(s)
- Anna S. Berghoff
- Department of Medicine I, Medical University of
Vienna, Vienna, Austria Comprehensive Cancer Center, Medical University of
Vienna, Vienna, Austria
| | - Matthias Preusser
- Department of Medicine I and Comprehensive
Cancer Center CNS Unit (CCC-CNS), Medical University of Vienna, Waehringer
Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
19
|
Sullivan RJ, Atkins MB, Kirkwood JM, Agarwala SS, Clark JI, Ernstoff MS, Fecher L, Gajewski TF, Gastman B, Lawson DH, Lutzky J, McDermott DF, Margolin KA, Mehnert JM, Pavlick AC, Richards JM, Rubin KM, Sharfman W, Silverstein S, Slingluff CL, Sondak VK, Tarhini AA, Thompson JA, Urba WJ, White RL, Whitman ED, Hodi FS, Kaufman HL. An update on the Society for Immunotherapy of Cancer consensus statement on tumor immunotherapy for the treatment of cutaneous melanoma: version 2.0. J Immunother Cancer 2018; 6:44. [PMID: 29848375 PMCID: PMC5977556 DOI: 10.1186/s40425-018-0362-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/17/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Cancer immunotherapy has been firmly established as a standard of care for patients with advanced and metastatic melanoma. Therapeutic outcomes in clinical trials have resulted in the approval of 11 new drugs and/or combination regimens for patients with melanoma. However, prospective data to support evidence-based clinical decisions with respect to the optimal schedule and sequencing of immunotherapy and targeted agents, how best to manage emerging toxicities and when to stop treatment are not yet available. METHODS To address this knowledge gap, the Society for Immunotherapy of Cancer (SITC) Melanoma Task Force developed a process for consensus recommendations for physicians treating patients with melanoma integrating evidence-based data, where available, with best expert consensus opinion. The initial consensus statement was published in 2013, and version 2.0 of this report is an update based on a recent meeting of the Task Force and extensive subsequent discussions on new agents, contemporary peer-reviewed literature and emerging clinical data. The Academy of Medicine (formerly Institute of Medicine) clinical practice guidelines were used as a basis for consensus development with an updated literature search for important studies published between 1992 and 2017 and supplemented, as appropriate, by recommendations from Task Force participants. RESULTS The Task Force considered patients with stage II-IV melanoma and here provide consensus recommendations for how they would incorporate the many immunotherapy options into clinical pathways for patients with cutaneous melanoma. CONCLUSION These clinical guidleines provide physicians and healthcare providers with consensus recommendations for managing melanoma patients electing treatment with tumor immunotherapy.
Collapse
Affiliation(s)
- Ryan J. Sullivan
- Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114 USA
| | | | | | - Sanjiv S. Agarwala
- St. Luke’s Cancer Center and Temple University, Center Valley, PA 18034 USA
| | | | | | | | | | | | | | - Jose Lutzky
- Mt. Sinai Medical Center, Miami Beach, FL 33140 USA
| | | | | | | | - Anna C. Pavlick
- New York University Cancer Institute, New York, NY 10016 USA
| | | | - Krista M. Rubin
- Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114 USA
| | - William Sharfman
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21231 USA
| | | | | | - Vernon K. Sondak
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612 USA
| | | | | | - Walter J. Urba
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR 97213 USA
| | | | | | | | - Howard L. Kaufman
- Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114 USA
| |
Collapse
|
20
|
Blake Z, Marks DK, Gartrell RD, Hart T, Horton P, Cheng SK, Taback B, Horst BA, Saenger YM. Complete intracranial response to talimogene laherparepvec (T-Vec), pembrolizumab and whole brain radiotherapy in a patient with melanoma brain metastases refractory to dual checkpoint-inhibition. J Immunother Cancer 2018; 6:25. [PMID: 29622046 PMCID: PMC5887256 DOI: 10.1186/s40425-018-0338-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 03/19/2018] [Indexed: 12/18/2022] Open
Abstract
Background Immunotherapy, in particular checkpoint blockade, has changed the clinical landscape of metastatic melanoma. Nonetheless, the majority of patients will either be primary refractory or progress over follow up. Management of patients progressing on first-line immunotherapy remains challenging. Expanded treatment options with combination immunotherapy has demonstrated efficacy in patients previously unresponsive to single agent or alternative combination therapy. Case presentation We describe the case of a patient with diffusely metastatic melanoma, including brain metastases, who, despite being treated with stereotactic radiosurgery and dual CTLA-4/PD-1 blockade (ipilimumab/nivolumab), developed systemic disease progression and innumerable brain metastases. This patient achieved a complete CNS response and partial systemic response with standard whole brain radiation therapy (WBRT) combined with Talimogene laherparepvec (T-Vec) and pembrolizumab. Conclusion Patients who do not respond to one immunotherapy combination may respond during treatment with an alternate combination, even in the presence of multiple brain metastases. Biomarkers are needed to assist clinicians in evidence based clinical decision making after progression on first line immunotherapy to determine whether response can be achieved with second line immunotherapy.
Collapse
Affiliation(s)
- Zoë Blake
- Columbia University Medical Center, Hematology/Oncology, 650 West 168th Street, New York, NY, 10032, USA
| | - Douglas K Marks
- NewYork-Prebsyterian/Columbia, Hematology/Oncology, 177 Fort Washington Avenue, New York, NY, 10032, USA
| | - Robyn D Gartrell
- Columbia University Medical Center, Hematology/Oncology, 650 West 168th Street, New York, NY, 10032, USA
| | - Thomas Hart
- Columbia University Medical Center, Hematology/Oncology, 650 West 168th Street, New York, NY, 10032, USA
| | - Patti Horton
- NewYork-Prebsyterian/Columbia, Hematology/Oncology, 161 Fort Washington Ave, New York, NY, 10032, USA
| | - Simon K Cheng
- NewYork-Prebsyterian/Columbia, Radiation Oncology, 161 Fort Washington Ave, New York, NY, 10032, USA
| | - Bret Taback
- NewYork-Prebsyterian/Columbia, Surgery, 161 Fort Washington Ave, New York, NY, 10032, USA
| | - Basil A Horst
- NewYork-Prebsyterian/Columbia, Dermatopathology, 630 W 168th Street, New York, NY, 10032, USA
| | - Yvonne M Saenger
- NewYork-Prebsyterian/Columbia, Hematology/Oncology, 161 Fort Washington Ave, New York, NY, 10032, USA.
| |
Collapse
|
21
|
The impact of timing of immunotherapy with cranial irradiation in melanoma patients with brain metastases: intracranial progression, survival and toxicity. J Neurooncol 2018; 138:299-306. [DOI: 10.1007/s11060-018-2795-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 02/01/2018] [Indexed: 10/18/2022]
|
22
|
Sun YW, Xu J, Zhou J, Liu WJ. Targeted drugs for systemic therapy of lung cancer with brain metastases. Oncotarget 2017; 9:5459-5472. [PMID: 29435193 PMCID: PMC5797064 DOI: 10.18632/oncotarget.23616] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/28/2017] [Indexed: 02/07/2023] Open
Abstract
Brain metastases are very common in lung cancer patients. The condition of these patients is complicated and difficult to treat, and adverse reactions following treatment can affect the nervous system, which severely reduces quality of life. Lung cancers are categorized as small cell lung cancers and non-small cell lung cancers. Patients with brain metastasis of small cell lung cancers are generally treated with brain radiotherapy and systemic chemotherapy, but stage III/IV patients with brain metastasis of non-small cell lung cancers are generally not responsive to radiotherapy or chemotherapy. With the recent development of targeted drugs, tumor molecular profile detection allows the selection of appropriate targeted drugs for adjuvant pharmacological treatment of brain metastasis in lung cancer patients. In recent years, immune checkpoint inhibitors have emerged and have been approved by the Food and Drug Administration (FDA) for the treatment of certain cancers, but their efficacy in lung cancer patients with brain metastases still needs to be confirmed. This paper focuses on highlighting drugs for targeted therapy of brain metastasis in lung cancer patients and their molecular targets and mechanisms of drug resistance.
Collapse
Affiliation(s)
- Ya-Wen Sun
- Shandong Cancer Hospital and Institute, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China
| | - Jian Xu
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.,Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Zhou
- University of South Carolina, Computer Science and Engineering Department, Columbia, SC, USA
| | - Wen-Juan Liu
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
23
|
Kamp MA, Fischer I, Dibué-Adjei M, Munoz-Bendix C, Cornelius JF, Steiger HJ, Slotty PJ, Turowski B, Rapp M, Sabel M. Predictors for a further local in-brain progression after re-craniotomy of locally recurrent cerebral metastases. Neurosurg Rev 2017; 41:813-823. [DOI: 10.1007/s10143-017-0931-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/01/2017] [Accepted: 11/20/2017] [Indexed: 01/01/2023]
|
24
|
Song JC, Ding XL, Sun XH, Safi M, Tian J. Brain metastasis in a patient with melanoma receiving Pembrolizumab therapy: A case report and review of the literature. Medicine (Baltimore) 2017; 96:e9278. [PMID: 29390382 PMCID: PMC5815794 DOI: 10.1097/md.0000000000009278] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 11/21/2017] [Accepted: 11/23/2017] [Indexed: 11/25/2022] Open
Abstract
RATIONALE Melanoma with brain metastasis is associated with a poor prognosis and high mortality rate. As patients with this condition have been excluded from most clinical trials, data on the use of anti-programmed death 1 therapy for these patients are limited. PATIENT CONCERNS The patient was a 62-year-old man with a 10-year history of melanotic nevus in his right forearm. He was admitted to another hospital in August 2015 due to the growth of the melanotic nevus over 1 year and complaint of a mass in the right mid-axillary area. The patient had no relevant medical, surgical, or family history. DIAGNOSES The biopsy of his right axillary lymph node showed malignant melanoma. INTERVENTIONS He was subsequently treated with adjuvant high-dose interferon after dacarbazine. Numerous metastatic lesions were found in his lung, abdomen, pelvic cavity, and brain after five months later, and then Pembrolizumab was used for six cycles (2 mg/kg every 3 weeks). He experienced immunorelated adverse events and we gave him cortisol to treat immunorelated disease until pneumonia was found. OUTCOMES We observed a delayed effect after three cycles of Pembrolizumab, the intracranial lesion presented clear margins and localization, while the other lesions became much smaller. A mixed response was observed after four cycles, with still stable extracranial metastases but growing a new lesion in brain. After two additional cycles of Pembrolizumab, the treatment was stopped due to the patient's inability to pay for it and a decline in his performance status. He then received palliative treatment at a local hospital and died for severe pulmonary infection, with an overall survival time of 7 months from metastasis. LESSONS In the case reported here, a delayed and mixed response was observed after Pembrolizumab was used. Because of causing severe pulmonary infection, the use of steroids should be considered carefully when treating immunorelated adverse events. It seemed that the Pembrolizumab has a positive effect on melanoma brain metastases especially combined with other treatments. However, there are still some challenges including patient selection, predictors of response, drug tolerance, optimizing combination strategies and control of adverse effects. More carefully designed clinical trials are urgently needed.
Collapse
|
25
|
Zhang Y, Liu X, Wang R, Liu S, Wang Y, Jing L, Louis MDJ, Cao R. Comparison of fusion protein and DC vaccine in inhibition of mouse B16F10 melanoma tumor. Biomed Pharmacother 2017; 97:784-792. [PMID: 29112931 DOI: 10.1016/j.biopha.2017.10.099] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/16/2017] [Accepted: 10/21/2017] [Indexed: 12/12/2022] Open
Abstract
Dendritic cell (DC) vaccine and fusion protein vaccine have been put into clinical use in cancer immunotherapy. This study compared DC vaccine and fusion protein vaccine directly in their capability of inducing specific immune response. We used mouse Granulocyte Macrophage-Colony Stimulating Factor (mGM-CSF) fused with gastrin-releasing peptide (GRP) and Gonadotrophin Releasing Hormone (GnRH) respectively to obtain mGM-CSF/GRP6 (mG6) and mGM-CSF/mGGn (mGGn) fusion proteins. We prepared fusion protein vaccine and DC vaccine including mG6 protein vaccine (6P), mGGn protein vaccine (nP), mG6 DC vaccine (6D) and mGGn DC vaccine (nD), then the two proteins were mixed to prepare combination proteins vaccine (6nP) and DC vaccine (6nD). After that, C57BL/6 mice were injected with B16F10 cell line to build melanoma tumor model, and were immunized with vaccines to produce antibodies to inhibit and destruct melanoma tumor cells. The discoveries showed that anti- mGM-CSF-GRP6 and anti- mGM-CSF-mGGn antibody vaccines were successfully created as expected; this was deduced from significant inhibition of melanoma tumor in vivo and significant reduction of tumor weight and volume. The effects of DC groups were better than that of the protein groups and the combination of vaccines were more effective than vaccine given separately. Our results indicate that using combination vaccine provides a new strategy to inhibit melanoma tumor growth but a complete cure of melanoma needs further investigations.
Collapse
Affiliation(s)
- Yan Zhang
- Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, TongjiaXiang 24, Nanjing, 210009, China
| | - Xiaoxin Liu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Tongjia Xiang 24, Nanjing, 210009, China
| | - Rui Wang
- Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, TongjiaXiang 24, Nanjing, 210009, China
| | - Shujun Liu
- Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, TongjiaXiang 24, Nanjing, 210009, China
| | - Yiqin Wang
- Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, TongjiaXiang 24, Nanjing, 210009, China
| | - Liangliang Jing
- Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, TongjiaXiang 24, Nanjing, 210009, China
| | | | - Rongyue Cao
- Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, TongjiaXiang 24, Nanjing, 210009, China.
| |
Collapse
|
26
|
Arozarena I, Wellbrock C. Overcoming resistance to BRAF inhibitors. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:387. [PMID: 29114545 PMCID: PMC5653517 DOI: 10.21037/atm.2017.06.09] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 05/25/2017] [Indexed: 12/14/2022]
Abstract
The discovery of activating mutations in the serine/threonine (S/T) kinase BRAF followed by a wave of follow-up research manifested that the MAPK-pathway plays a critical role in melanoma initiation and progression. BRAF and MEK inhibitors produce an unparalleled response rate in melanoma, but it is now clear that most responses are transient, and while some patients show long lasting responses the majority progress within 1 year. In accordance with the key role played by the MAPK-pathway in BRAF mutant melanomas, disease progression is mostly due to the appearance of drug-resistance mechanisms leading to restoration of MAPK-pathway activity. In the present article we will review the development, application and clinical effects of BRAF and MEK inhibitors both, as single agent and in combination in the context of targeted therapy in melanoma. We will then describe the most prominent mechanisms of resistance found in patients progressed on these targeted therapies. Finally we will discuss strategies for further optimizing the use of MAPK inhibitors and will describe the potential of alternative combination therapies to either delay the onset of resistance to MAPK inhibitors or directly target specific mechanisms of resistance to BRAF/MEK inhibitors.
Collapse
Affiliation(s)
- Imanol Arozarena
- Navarrabiomed-Fundación Miguel Servet-Idisna, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Claudia Wellbrock
- Manchester Cancer Research Centre, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
27
|
Nguyen SM, Castrellon A, Vaidis O, Johnson AE. Stereotactic Radiosurgery and Ipilimumab Versus Stereotactic Radiosurgery Alone in Melanoma Brain Metastases. Cureus 2017; 9:e1511. [PMID: 28959506 PMCID: PMC5612568 DOI: 10.7759/cureus.1511] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Benefits of stereotactic radiosurgery (SRS) have been well established in melanoma brain metastases (MBM). Immunotherapy agents such as ipilimumab (ipi) have recently demonstrated clinical efficacy in advanced disease as well. The theoretical synergistic effects of combining these therapies in MBM have not been explored in detail, however, we conducted a systematic review with meta-analysis of studies that compared combined SRS and ipi versus SRS alone in MBM. Medical Literature Analysis and Retrieval System Online (MEDLINE) and Central databases were used for our literature search, which was conducted by three reviewers. We included studies that examined SRS and ipilimumab compared to SRS alone in MBM. Pertinent results were tabulated in a standardized spreadsheet. Newcastle-Ottawa Scale (NOS) Risk of Bias Assessment and Grading of Recommendations, Assessment, Development and Evaluation (GRADE) method for rating evidence quality were used for qualitative analysis. Review Manager was used for statistical analysis. We identified four cohort studies that compared SRS plus ipi versus SRS alone in MBM. As per the GRADE criteria, we found low-quality evidence for survival benefits associated with combined treatment. Meta-analysis confirmed a significant benefit in survival for SRS and ipilimumab (hazard ratio 0.38, 95% confidence interval 0.28 – 0.52, p < 0.01). There were no significant differences between comparison groups for local control, distant brain control, radiation necrosis, or intracranial bleeding. We conclude that low-quality evidence exists for superior overall survival in MBM treated with SRS and ipilimumab compared to SRS without ipilimumab. There is also no increased risk of radiation necrosis and/or intracranial bleeding with combining radiation and immunotherapy in this setting.
Collapse
Affiliation(s)
| | | | - Oliver Vaidis
- Department of Mathematics and Statistics, University of South Florida
| | | |
Collapse
|