1
|
Söylemez CM, Gürsoy P, Şanli UA. Response rates of pazopanib therapy in metastatic soft tissue sarcoma using real‑world data. Oncol Lett 2025; 29:102. [PMID: 39736927 PMCID: PMC11683527 DOI: 10.3892/ol.2024.14848] [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: 04/23/2024] [Accepted: 09/23/2024] [Indexed: 01/01/2025] Open
Abstract
The present study was a retrospective single-center study. A total of 81 patients diagnosed with metastatic soft tissue sarcoma were included who received pazopanib therapy. Clinical data, including age at diagnosis, histological subtype, treatments received before pazopanib, number of metastatic sites at the time of initiation of treatment, progression-free survival and overall survival time under pazopanib treatment, side effects and response evaluation in follow-up imaging after initiation of pazopanib therapy, were recorded. The 81 patients had 11 different histological subtypes. The synovial sarcoma, leiomyosarcoma and pleomorphic sarcoma groups included 51 patients in total. The median overall survival time in the entire study cohort was 46 months, and the median progression-free survival time was 5 months. The clinical response rate was 46.3%. Patients with hemangioendothelioma and alveolar soft part sarcoma exhibited an improved response to treatment compared with that of patients with other subtypes. Line of therapy and tumor grade were not significantly associated with progression-free survival or clinical response. It was concluded that, regardless of subtype, patients with a low tumor grade and a small number of metastatic sites exhibited an improved response; although the difference in response for patients with a low tumor grade was not significant. In addition, administering the treatment as a second- or third-line therapy appeared to be more appropriate compared with administering it as a later-line therapy; however, this difference was not found to be statistically significant. Therefore, pazopanib should be evaluated as an option for a selected group of patients in whom these factors present together. A further advantage of pazopanib demonstrated was that treatment tolerance was generally good.
Collapse
Affiliation(s)
- Cem Murat Söylemez
- Department of Medical Oncology, University of Health Science Izmir Tepecik Research and Training Hospital, Bornova, Izmir 35110, Turkey
| | - Pinar Gürsoy
- Department of Medical Oncology, Ege University Faculty of Medicine, Bornova, Izmir 35100, Turkey
| | - Ulus Ali Şanli
- Department of Medical Oncology, Ege University Faculty of Medicine, Bornova, Izmir 35100, Turkey
| |
Collapse
|
2
|
Matsuoka H, Yoshida KI, Nakai S, Suzuki R, Imura Y, Takami H, Watanabe M, Wakamatsu T, Tamiya H, Outani H, Yagi T, Kakunaga S, Takenaka S. Successful pazopanib treatment of undifferentiated pleomorphic sarcoma with coamplification of PDGFRA, VEGFR2 and KIT: A case report. Mol Clin Oncol 2024; 21:69. [PMID: 39113850 PMCID: PMC11304161 DOI: 10.3892/mco.2024.2767] [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: 02/06/2024] [Accepted: 06/03/2024] [Indexed: 08/10/2024] Open
Abstract
Undifferentiated pleomorphic sarcoma (UPS) is a high-grade, aggressive soft tissue sarcoma (STS) with a poor prognosis, and no definitive or effective treatment is currently available for it. Pazopanib, an orally available multiple tyrosine kinase inhibitor, has been approved for the treatment of advanced STS. The present study documents the case of a 51-year-old man with advanced UPS with coamplification of platelet-derived growth factor receptor A (PDGFRA), vascular endothelial growth factor receptor 2 (VEGFR2) and stem cell factor receptor (KIT) genes. The patient exhibited a marked and sustained response to pazopanib. The patient presented with a retroperitoneal tumour with pancreatic head lymph node metastasis, and bone metastases in the second/fifth thoracic vertebrae and left femur. Based on the histological analysis of the retroperitoneal tumour and femoral mass, the patient was diagnosed with UPS. Palliative radiation therapy was administered to the left femur and second/fifth thoracic vertebrae to prevent fractures. After radiation therapy, the patient achieved a partial response after eight courses of doxorubicin. A comprehensive genomic profiling analysis (FoundationOne® CDx) revealed coamplification of PDGFRA, VEGFR2 and KIT genes. Hence, pazopanib was initiated as a second-line treatment. Notably, the retroperitoneal tumour shrank, and no new lesions developed for 3 years after the initiation of pazopanib treatment. This response suggests that the coamplification of PDGFRA, VEGFR2 and KIT may predict favourable outcomes in response to pazopanib.
Collapse
Affiliation(s)
- Haruki Matsuoka
- Musculoskeletal Oncology Service, Osaka International Cancer Institute, Osaka 541-8567, Japan
- Department of Orthopaedic Surgery, Osaka Police Hospital, Osaka 543-0035, Japan
| | - Ken-Ichi Yoshida
- Department of Diagnostic Pathology and Cytology, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Sho Nakai
- Musculoskeletal Oncology Service, Osaka International Cancer Institute, Osaka 541-8567, Japan
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Rie Suzuki
- Musculoskeletal Oncology Service, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Yoshinori Imura
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Haruna Takami
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Makiyo Watanabe
- Musculoskeletal Oncology Service, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Toru Wakamatsu
- Musculoskeletal Oncology Service, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Hironari Tamiya
- Musculoskeletal Oncology Service, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Hidetatsu Outani
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - Toshinari Yagi
- Musculoskeletal Oncology Service, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Shigeki Kakunaga
- Musculoskeletal Oncology Service, Osaka International Cancer Institute, Osaka 541-8567, Japan
| | - Satoshi Takenaka
- Musculoskeletal Oncology Service, Osaka International Cancer Institute, Osaka 541-8567, Japan
| |
Collapse
|
3
|
Arfan S, Thway K, Jones RL, Huang PH. Molecular Heterogeneity in Leiomyosarcoma and Implications for Personalised Medicine. Curr Treat Options Oncol 2024; 25:644-658. [PMID: 38656686 DOI: 10.1007/s11864-024-01204-5] [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] [Accepted: 04/04/2024] [Indexed: 04/26/2024]
Abstract
OPINION STATEMENT Leiomyosarcoma (LMS) is one of the more common subtypes of soft tissue sarcomas (STS), accounting for about 20% of cases. Differences in anatomical location, risk of recurrence and histomorphological variants contribute to the substantial clinical heterogeneity in survival outcomes and therapy responses observed in patients. There is therefore a need to move away from the current one-size-fits-all treatment approach towards a personalised strategy tailored for individual patients. Over the past decade, tissue profiling studies have revealed key genomic features and an additional layer of molecular heterogeneity among patients, with potential utility for optimal risk stratification and biomarker-matched therapies. Furthermore, recent studies investigating intratumour heterogeneity and tumour evolution patterns in LMS suggest some key features that may need to be taken into consideration when designing treatment strategies and clinical trials. Moving forward, national and international collaborative efforts to aggregate expertise, data, resources and tools are needed to achieve a step change in improving patient survival outcomes in this disease of unmet need.
Collapse
Affiliation(s)
- Sara Arfan
- Division of Molecular Pathology, The Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
| | - Khin Thway
- Division of Molecular Pathology, The Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
- The Royal Marsden NHS Foundation Trust, Fulham Road, London, SW3 6JJ, UK
| | - Robin L Jones
- The Royal Marsden NHS Foundation Trust, Fulham Road, London, SW3 6JJ, UK
- Division of Clinical Studies, The Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK
| | - Paul H Huang
- Division of Molecular Pathology, The Institute of Cancer Research, 15 Cotswold Road, Sutton, SM2 5NG, UK.
| |
Collapse
|
4
|
Kato S, Gumas S, Adashek JJ, Okamura R, Lee S, Sicklick JK, Kurzrock R. Multi-omic analysis in carcinoma of unknown primary (CUP): therapeutic impact of knowing the unknown. Mol Oncol 2024; 18:956-968. [PMID: 35866362 PMCID: PMC10994241 DOI: 10.1002/1878-0261.13293] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 05/11/2022] [Accepted: 07/20/2022] [Indexed: 11/05/2022] Open
Abstract
Carcinoma of unknown primary (CUP) is a difficult-to-manage malignancy. Multi-omic profiles and treatment outcome vs. degree of precision matching were assessed. Tumours underwent next-generation sequencing (NGS) [tissue and/or blood-derived cell-free DNA (cfDNA)]. Selected patients had transcriptome-based immune profiling and/or programmed cell death 1 ligand 1 (PD-L1) immunohistochemistry analysis. Patients could be reviewed by a Molecular Tumor Board, but physicians chose the therapy. Of 6497 patients in the precision database, 97 had CUP. The median number of pathogenic tissue genomic alterations was 4 (range, 0-25), and for cfDNA, was 2 (range, 0-9). Each patient had a distinct molecular landscape. Food and Drug Administration (FDA)-approved biomarkers included the following: PD-L1+ ≥ 1%, 30.9% of CUPs tested; microsatellite instability, 3.6%; tumour mutational burden ≥ 10 mutations·Mb-1, 23%; and neurotrophic receptor tyrosine kinase (NTRK) fusions, 0%. RNA-based immunograms showed theoretically druggable targets: lymphocyte activation gene 3 protein (LAG-3), macrophage colony-stimulating factor 1 receptor (CSF1R), adenosine receptor A2 (ADORA2) and indoleamine 2,3-dioxygenase 1 (IDO1). Overall, 56% of patients had ≥ 1 actionable biomarker (OncoKB database). To quantify the degree of matching (tumours to drugs), a Matching Score (MS; roughly equivalent to number of alterations targeted/total number of deleterious alterations) was calculated post hoc. Comparing evaluable treated patients [MS high, > 50% (N = 15) vs. low ≤ 50% (N = 47)], median progression-free survival was 10.4 vs. 2.8 months (95% CI 0.11-0.64; HR 0.27; P = 0.002); survival, 15.8 vs. 6.9 months (95% CI 0.17-1.16; HR 0.45; P = 0.09); and clinical benefit rate (stable disease ≥ 6 months/partial/complete response), 71% vs. 24% (P = 0.003). Higher MS was the only factor that predicted improvement in outcome variables after multivariate analysis. In conclusion, CUPs are molecularly complex. Treatments with high degrees of matching to molecular alterations (generally achieved by individualized combinations) correlated with improved outcomes.
Collapse
Affiliation(s)
- Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of MedicineUC San Diego Moores Cancer CenterLa JollaCAUSA
| | - Sophia Gumas
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of MedicineUC San Diego Moores Cancer CenterLa JollaCAUSA
| | - Jacob J. Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer CenterThe Johns Hopkins HospitalBaltimoreMDUSA
| | | | - Suzanna Lee
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of MedicineUC San Diego Moores Cancer CenterLa JollaCAUSA
| | - Jason K. Sicklick
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of MedicineUC San Diego Moores Cancer CenterLa JollaCAUSA
- Division of Surgical Oncology, Department of SurgeryUC San Diego School of MedicineCAUSA
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of MedicineUC San Diego Moores Cancer CenterLa JollaCAUSA
| |
Collapse
|
5
|
Mochizuki T, Ikegami M, Akiyama T. Factors predictive of second-line chemotherapy in soft tissue sarcoma: An analysis of the National Genomic Profiling Database. Cancer Sci 2024; 115:575-588. [PMID: 38115234 PMCID: PMC10859616 DOI: 10.1111/cas.16050] [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: 10/17/2023] [Revised: 11/28/2023] [Accepted: 12/06/2023] [Indexed: 12/21/2023] Open
Abstract
Of the drugs used in second-line chemotherapy for soft tissue sarcoma (STS), trabectedin is effective for liposarcoma and leiomyosarcoma (L-sarcoma), eribulin for liposarcoma, and pazopanib for non-liposarcoma. The indications for these drugs in STS other than L-sarcoma have not been established. Here we explored the prognosis, mutation profiles, and drug-response factors in STS using real-world big data. Clinicogenomic data on 1761 patients with sarcoma who underwent FoundationOne CDx were obtained from a national database in Japan. Patients with TP53 and KDM2D mutations had a significantly shorter survival period of 253 (95% CI, 99-404) and 330 (95% CI, 20-552) days, respectively, than those without mutations. Non-supervised clustering based on mutation profiles generated 13 tumor clusters. The response rate (RR) to trabectedin was highest in an MDM2-amplification cluster (odds ratio [OR]: 2.2; p = 0.2). The RR was lowest for eribulin in an MDM2-amplification cluster (OR: 0.4; p = 0.03) and highest in a TERT-mutation cluster (OR: 3.0; p = 0.03). The RR was highest for pazopanib in a PIK3CA/PTEN-wild type cluster (OR: 2.1; p = 0.03). In particular, patients harboring mutations in genes regulating the PI3K/Akt/mTOR pathway had a lower RR than patients without mutations (OR: 0.3; p = 0.04). In STS, mutation profiles were more useful in predicting the drug response than histology. The present study demonstrated the potential of tailored therapy guided by mutation profiles established by comprehensive genomic profiling testing in optimizing second-line chemotherapy for STS. The findings of this study will hopefully contribute some valuable insights into enhancing STS treatment strategies and outcomes.
Collapse
Affiliation(s)
- Takao Mochizuki
- Department of Orthopaedic Surgery, Saitama Medical CenterJichi Medical UniversitySaitamaJapan
- Department of Musculoskeletal OncologyTokyo Metropolitan Cancer and Infectious Diseases Center, Komagome HospitalTokyoJapan
| | - Masachika Ikegami
- Department of Musculoskeletal OncologyTokyo Metropolitan Cancer and Infectious Diseases Center, Komagome HospitalTokyoJapan
- Division of Cellular SignalingNational Cancer Center Research InstituteTokyoJapan
| | - Toru Akiyama
- Department of Orthopaedic Surgery, Saitama Medical CenterJichi Medical UniversitySaitamaJapan
| |
Collapse
|
6
|
Pan M, Zhou MY, Jiang C, Zhang Z, Bui N, Bien J, Siy A, Achacoso N, Solorzano AV, Tse P, Chung E, Hu W, Thomas S, Ganjoo K, Habel LA. PTEN pathogenic variants are associated with poor prognosis in patients with advanced soft tissue sarcoma. BJC REPORTS 2024; 2:9. [PMID: 39516677 PMCID: PMC11524139 DOI: 10.1038/s44276-023-00029-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 11/16/2024]
Abstract
BACKGROUND We aimed to examine whether PTEN pathogenic variants (mutPTEN) were associated with overall survival (OS) in patients with advanced soft tissue sarcoma (STS) with the presence of one or more of the most common genomic alterations including p53, CDKN2A, RB1, and ATRX pathogenic variants. METHODS This study included patients from Kaiser Permanente Northern California and Stanford Cancer Center with grade 2 or higher locally advanced and metastatic STS. RESULTS A total of 174 patients had leiomyosarcoma (LMS), 136 had undifferentiated pleomorphic sarcoma (UPS), 78 had Liposarcoma (LPS), and 214 had other histology subtypes (Others). Among all patients with STS, OS was worse for those with mutPTEN versus wild-type PTEN (wtPTEN, adjusted HR [aHR] = 1.58 [95% CI, 1.11-2.23]), mutCDKN2A vs wtCDKN2A (aHR = 1.33 [95% CI .99-1.80]), and mutRB1 vs wtRB1 (aHR = 1.26 [95% CI 0.93-1.70[), while OS was similar for mutp53 vs wtp53 and mutATRX vs wtATRX. MutPTEN versus wtPTEN was consistently associated with worse OS in histologic subtypes including LMS and UPS and molecular subgroups. CONCLUSION MutPTEN vs wtPTEN was associated with worse OS in advanced STS. If confirmed, our findings could be helpful for prognostic stratification in clinical practice and for further understanding the molecular mechanisms of STS.
Collapse
Affiliation(s)
- Minggui Pan
- Division of Research, Kaiser Permanente, Oakland, CA, 94612, USA.
- Sarcoma Program, Division of Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Maggie Y Zhou
- Sarcoma Program, Division of Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Chen Jiang
- Division of Research, Kaiser Permanente, Oakland, CA, 94612, USA
| | - Zheyang Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Nam Bui
- Sarcoma Program, Division of Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jeffrey Bien
- Sarcoma Program, Division of Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Amanda Siy
- Sarcoma Program, Division of Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Ninah Achacoso
- Division of Research, Kaiser Permanente, Oakland, CA, 94612, USA
| | | | - Pam Tse
- Division of Research, Kaiser Permanente, Oakland, CA, 94612, USA
| | - Elaine Chung
- Division of Research, Kaiser Permanente, Oakland, CA, 94612, USA
| | - Wenwei Hu
- Rutger's Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA
| | - Sachdev Thomas
- Department of Oncology and Hematology, Kaiser Permanente, Vallejo, CA, 94589, USA
| | - Kristen Ganjoo
- Sarcoma Program, Division of Oncology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Laurel A Habel
- Division of Research, Kaiser Permanente, Oakland, CA, 94612, USA
| |
Collapse
|
7
|
Stacchiotti S, Baldi GG, Frezza AM, Morosi C, Greco FG, Collini P, Barisella M, Dagrada GP, Zaffaroni N, Pasquali S, Gronchi A, Huang P, Ingrosso M, Tinè G, Miceli R, Casali PG. Regorafenib in advanced solitary fibrous tumour: Results from an exploratory phase II clinical study. Eur J Cancer 2023; 195:113391. [PMID: 37918286 DOI: 10.1016/j.ejca.2023.113391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/29/2023] [Accepted: 10/12/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND To investigate the activity of regorafenib in advanced solitary fibrous tumour (SFT). METHODS An Italian monocentric investigator-initiated exploratory single-arm Phase II trial was conducted of regorafenib in adult patients with advanced and progressive SFT, until progression or limiting toxicity. Prior treatment with antiangiogenics was allowed. Primary and secondary end-points were: overall response rate (ORR) by Choi criteria, and ORR by RECIST, progression-free survival (PFS), overall survival (OS). RESULTS From January 2016 to February 2021, 18 patients were enroled [malignant-SFT = 13; dedifferentiated-SFT (D-SFT) = 4; typical-SFT (T-SFT) = 1]. Fourteen patients were pre-treated, in 12 cases with antiangiogenics (median [m-] lines of treatment = 3). Sixteen patients were evaluable for response (one screening failure; one early discontinuation). Six/16 (35.2%) required a definitive dose reduction. ORR by Choi was 37.5% (95% CI: 15.2-64.6), with 6/16 (37.5%) partial responses (PR), 6/16 (37.5%) stable disease (SD) and 4/16 (25%) progressions; 5/6 responses occurred in patients pre-treated with antiangiogenics. No responses were detected in D-SFT. Best RECIST responses were: 1/16 (6.2%) PR, 12/16 (75%) SD, 3/16 (18.8%) progressions. At 48.4 month m-FU, m-PFS by Choi was 4.7 (inter-quartile range: 2.4-13.1) months, with 31.2% patients progression-free at 1 year. CONCLUSION Regorafenib showed activity in SFT, with 30% patients free-from-progression at one year. Responses were observed also in patients pretreated and refractory to another antiangiogenic agents. However, ORR and m-PFS were lower than reported with other antiangiogenics, and this was possibly due to discrepancies in the patient population and the high-rate of dose reductions.
Collapse
Affiliation(s)
- Silvia Stacchiotti
- Medical Oncology Unit 2, Cancer Medicine Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale Tumori, Milan, Italy.
| | | | - Anna Maria Frezza
- Medical Oncology Unit 2, Cancer Medicine Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale Tumori, Milan, Italy
| | - Carlo Morosi
- Department of Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Paola Collini
- Soft Tissue Tumor Pathology Unit, Advanced Diagnostics Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marta Barisella
- Soft Tissue Tumor Pathology Unit, Advanced Diagnostics Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Gian Paolo Dagrada
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Nadia Zaffaroni
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sandro Pasquali
- Molecular Pharmacology Unit, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessandro Gronchi
- Sarcoma Unit, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Paul Huang
- Division of Molecular Pathology, Institute of Cancer Research, London, UK
| | - Matilde Ingrosso
- Medical Oncology Unit 2, Cancer Medicine Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale Tumori, Milan, Italy
| | - Gabriele Tinè
- Unit of Biostatistics for Clinical Research, Department Epidemiology and Data Science, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Rosalba Miceli
- Unit of Biostatistics for Clinical Research, Department Epidemiology and Data Science, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Paolo Giovanni Casali
- Medical Oncology Unit 2, Cancer Medicine Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale Tumori, Milan, Italy
| |
Collapse
|
8
|
Nishio M, Paz-Ares L, Reck M, Nakagawa K, Garon EB, Popat S, Ceccarelli M, Graham HT, Visseren-Grul C, Novello S. RELAY, Ramucirumab plus Erlotinib (RAM+ERL) in Untreated Metastatic EGFR-Mutant NSCLC (EGFR+ NSCLC): Association between TP53 Status and Clinical Outcome. Clin Lung Cancer 2023:S1525-7304(23)00046-3. [PMID: 37076395 DOI: 10.1016/j.cllc.2023.02.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 04/03/2023]
Abstract
BACKGROUND Ramucirumab plus erlotinib (RAM+ERL) demonstrated superior progression-free survival (PFS) in RELAY, a randomised Phase III trial in patients with untreated, metastatic, EGFR-mutated, non-small-cell lung cancer (EGFR+ NSCLC). Here, we present the relationship between TP53 status and outcomes in RELAY. MATERIALS AND METHODS Patients received oral ERL plus intravenous RAM (10 mg/kg IV) or placebo (PBO+ERL) every 2 weeks. Plasma was assessed by Guardant 360 next-generation sequencing and patients with any gene alteration detected at baseline were included in this exploratory analysis. Endpoints included PFS, overall response rate (ORR), disease control rate (DCR), DoR, overall survival (OS), safety, and biomarker analysis. The association between TP53 status and outcomes was evaluated. RESULTS Mutated TP53 was detected in 165 (42.7%; 74 RAM+ERL, 91 PBO+ERL) patients, wild-type TP53 in 221 (57.3%; 118 RAM+ERL, 103 PBO+ERL) patients. Patient and disease characteristics and concurrent gene alterations were comparable between those with mutant and wildtype TP53. Independent of treatment, TP53 mutations, most notably on exon 8, were associated with worse clinical outcomes. In all patients, RAM+ERL improved PFS. While ORR and DCR were comparable across all patients, DoR was superior with RAM+ERL. There were no clinically meaningful differences in the safety profiles between those with baseline TP53 mutation and wild-type. CONCLUSION This analysis indicates that while TP53 mutations are a negative prognostic marker in EGFR+ NSCLC, the addition of a VEGF inhibitor improves outcomes in those with mutant TP53. RAM+ERL is an efficacious first-line treatment option for patients with EGFR+ NSCLC, independent of TP53 status.
Collapse
Affiliation(s)
- Makoto Nishio
- Department of Thoracic Medical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Luis Paz-Ares
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Martin Reck
- Department of Thoracic Oncology, LungenClinic, Airway Research Center North, German Center for Lung Research, Grosshansdorf, Germany
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Edward B Garon
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Sanjay Popat
- Lung Unit, Royal Marsden NHS Trust, London, United Kingdom
| | - Matteo Ceccarelli
- Global Clinical Development, Eli Lilly and Company, Sesto Fiorentino, Florence, Italy
| | | | - Carla Visseren-Grul
- Global Clinical Development, Eli Lilly Netherlands, Utrecht, The Netherlands
| | - Silvia Novello
- Department of Oncology, University of Turin, San Luigi Hospital, Turin, Italy
| |
Collapse
|
9
|
Chen X, Liu T, Wu J, Zhu C, Guan G, Zou C, Guo Q, Ren X, Li C, Cheng P, Cheng W, Wu A. Molecular profiling identifies distinct subtypes across TP53 mutant tumors. JCI Insight 2022; 7:156485. [PMID: 36256461 PMCID: PMC9746906 DOI: 10.1172/jci.insight.156485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 10/14/2022] [Indexed: 01/12/2023] Open
Abstract
Tumor protein 53 mutation (TP53mut) is one of the most important driver events facilitating tumorigenesis, which could induce a series of chain reactions to promote tumor malignant transformation. However, the malignancy progression patterns under TP53 mutation remain less known. Clarifying the molecular landscapes of TP53mut tumors will help us understand the process of tumor development and aid precise treatment. Here, we distilled genetic and epigenetic features altered in TP53mut cancers for cluster-of-clusters analysis. Using integrated classification, we derived 5 different subtypes of TP53mut patients. These subtypes have distinct features in genomic alteration, clinical relevance, microenvironment dysregulation, and potential therapeutics. Among the 5 subtypes, COCA3 was identified as the subtype with worst prognosis, causing an immunosuppressive microenvironment and immunotherapeutic resistance. Further drug efficacy research highlighted olaparib as the most promising therapeutic agents for COCA3 tumors. Importantly, the therapeutic efficacy of olaparib in COCA3 and immunotherapy in non-COCA3 tumors was validated via in vivo experimentation. Our study explored the important molecular events and developed a subtype classification system with distinct targeted therapy strategies for different subtypes of TP53mut tumors. These multiomics classification systems provide a valuable resource that significantly expands the knowledge of TP53mut tumors and may eventually benefit in clinical practice.
Collapse
Affiliation(s)
- Xin Chen
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Tianqi Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jianqi Wu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chen Zhu
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Gefei Guan
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Cunyi Zou
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qing Guo
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaolin Ren
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China.,Department of Neurosurgery, Shenyang Red Cross Hospital, Shenyang, Liaoning, China
| | - Chen Li
- Department of Orthodontics, Stomatological Hospital of China Medical University, Shenyang, Liaoning, China
| | - Peng Cheng
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wen Cheng
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.,Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Anhua Wu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.,Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
10
|
Napolitano A, Moura DS, Hindi N, Mondaza-Hernandez JL, Merino-Garcia JA, Ramos R, Dagrada GP, Stacchiotti S, Graziano F, Vincenzi B, Martin-Broto J. Expression of p53 as a biomarker of pazopanib efficacy in solitary fibrous tumours: translational analysis of a phase II trial. Ther Adv Med Oncol 2022; 14:17588359221116155. [PMID: 35965642 PMCID: PMC9364178 DOI: 10.1177/17588359221116155] [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: 10/11/2021] [Accepted: 07/11/2022] [Indexed: 11/28/2022] Open
Abstract
Background: Solitary fibrous tumours (SFT) are soft tissue sarcomas molecularly defined
by the presence of the NAB2::STAT6 intrachromosomal fusion gene. Recently, a
prospective phase II trial evaluating the role of the antiangiogenic
tyrosine kinase inhibitor pazopanib in SFT has been conducted
(NCT02066285). Methods: Here, we analysed the mRNA and protein expression levels of the tumour
suppressor and angiogenesis regulator p53 (TP53) in
pre-treatment tumour samples from 22 patients with low aggressive (or
typical) SFT and 28 patients with high aggressive (26 malignant and 2
dedifferentiated) SFT enrolled in the aforementioned pazopanib phase II
trial. These results were correlated with radiological progression-free
survival (PFS) and objective response. Univariate and multivariate Cox
regression analyses were also performed, including known clinic-pathological
prognostic factors. Results: Diffuse immunohistochemistry (IHC) expression of p53 was only found in
patients with aggressive SFT and was associated with significantly shorter
PFS [hazard ratio (HR): 4.39, 95% confidence interval (CI): 1.19–16.14).
TP53 mRNA levels were significantly higher in the low
aggressive SFT group. Only in the high aggressive SFT group, relatively
higher levels of TP53 were significantly associated with
shorter PFS (HR: 4.16, 95% CI: 1.46–11.89) as well as to a lower rate of
disease control following treatment with pazopanib. In the multivariate
analysis, the only independent prognostic factor in the whole cohort was
mitotic count. Conclusion: Diffuse p53 IHC expression and higher TP53 mRNA levels are
associated with worse prognosis in the subset of aggressive SFT patients
treated with pazopanib.
Collapse
Affiliation(s)
- Andrea Napolitano
- Department of Medical Oncology, University Campus Bio-Medico, Rome, Italy
| | - David S Moura
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid (IIS/FJD-UAM), Madrid, Spain
| | - Nadia Hindi
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid (IIS/FJD-UAM), Madrid, Spain
| | - José L Mondaza-Hernandez
- Health Research Institute Fundacion Jimenez Diaz, Universidad Autonoma de Madrid (IIS/FJD-UAM), Madrid, Spain
| | - José A Merino-Garcia
- Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rafael Ramos
- Department of Pathology, University Hospital Son Espases, Palma, Spain
| | - Gian Paolo Dagrada
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvia Stacchiotti
- Department of Cancer Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Bruno Vincenzi
- Department of Medical Oncology, University Campus Bio-Medico, via Alvaro del Portillo 200, Rome 00128, Italy
| | | |
Collapse
|
11
|
Bondili SK, Nandhana R, Dhanawat A, Noronha V, Joshi A, Patil VM, Menon N, Kaushal RK, Choughule A, Jiwnani SS, Janu A, Prabhash K. Characteristics and clinical outcomes of pulmonary sarcomatoid carcinoma: experience from Tata Memorial Centre. Ecancermedicalscience 2022; 16:1438. [PMID: 36200011 PMCID: PMC9470174 DOI: 10.3332/ecancer.2022.1438] [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: 02/02/2022] [Indexed: 11/06/2022] Open
Abstract
Background Pulmonary sarcomatoid carcinoma (PSC) constitutes a heterogeneous group of poorly differentiated non-small cell lung cancers. Since these are rare tumours, we sought to determine the characteristics and clinical outcomes of these patients treated at our centre. Methods We did a retrospective evaluation of all patients diagnosed with PSC between January 2013 and September 2020 at the Tata Memorial Hospital, Mumbai, India. Baseline demographic and treatment data and outcomes were obtained retrospectively from electronic medical records and survival was calculated by using the Kaplan-Meier method. Results Out of 151 patients diagnosed with PSC during this period, 129 were included in the final analysis. The clinical stage was stage I in 3 (2.03%), stage II in 4 (3.1%), stage III in 35 (27.1%) and stage IV in 87 (67.4%). The median follow-up duration was 32 months (range, 15.0-48.9). The median overall survival (OS) of patients who received curative surgery was 18 months (95% confidence interval (95% CI), 2.59-33.4); concurrent chemoradiation was 11 months (95% CI, 2.99-19); palliative chemotherapy was 8 months (95% CI, 5.24-10.75) and best supportive care was 1 month (95% CI, 0.43-1.57, p = 0.001). On multivariate analysis, the presence of brain metastasis (p = 0.018; hazard ratio (HR), 2.47; 95% CI, 1.34-4.49) and the administration of chemotherapy (p = 0.037; HR, 2.2; 95% CI, 1.04-4.94) were the only factors impacting the OS. Conclusion PSC usually presents in advanced stages and is associated with a poor prognosis.
Collapse
Affiliation(s)
| | | | | | - Vanita Noronha
- Tata Memorial Hospital, Mumbai, Maharashtra, 400 012, India
| | - Amit Joshi
- Tata Memorial Hospital, Mumbai, Maharashtra, 400 012, India
| | | | - Nandini Menon
- Tata Memorial Hospital, Mumbai, Maharashtra, 400 012, India
| | | | | | | | - Amit Janu
- Tata Memorial Hospital, Mumbai, Maharashtra, 400 012, India
| | - Kumar Prabhash
- Tata Memorial Hospital, Mumbai, Maharashtra, 400 012, India
| |
Collapse
|
12
|
Heilig CE, Laßmann A, Mughal SS, Mock A, Pirmann S, Teleanu V, Renner M, Andresen C, Köhler BC, Aybey B, Bauer S, Siveke JT, Hamacher R, Folprecht G, Richter S, Schröck E, Brandts CH, Ahrens M, Hohenberger P, Egerer G, Kindler T, Boerries M, Illert AL, von Bubnoff N, Apostolidis L, Jost PJ, Westphalen CB, Weichert W, Keilholz U, Klauschen F, Beck K, Winter U, Richter D, Möhrmann L, Bitzer M, Schulze-Osthoff K, Brors B, Mechtersheimer G, Kreutzfeldt S, Heining C, Lipka DB, Stenzinger A, Schlenk RF, Horak P, Glimm H, Hübschmann D, Fröhling S. Gene expression-based prediction of pazopanib efficacy in sarcoma. Eur J Cancer 2022; 172:107-118. [PMID: 35763870 DOI: 10.1016/j.ejca.2022.05.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/28/2022] [Accepted: 05/12/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND The multi-receptor tyrosine kinase inhibitor pazopanib is approved for the treatment of advanced soft-tissue sarcoma and has also shown activity in other sarcoma subtypes. However, its clinical efficacy is highly variable, and no reliable predictors exist to select patients who are likely to benefit from this drug. PATIENTS AND METHODS We analysed the molecular profiles and clinical outcomes of patients with pazopanib-treated sarcoma enrolled in a prospective observational study by the German Cancer Consortium, DKTK MASTER, that employs whole-genome/exome sequencing and transcriptome sequencing to inform the care of young adults with advanced cancer across histology and patients with rare cancers. RESULTS Among 109 patients with available whole-genome/exome sequencing data, there was no correlation between clinical parameters, specific genetic alterations or mutational signatures and clinical outcome. In contrast, the analysis of a subcohort of 62 patients who underwent molecular analysis before pazopanib treatment and had transcriptome sequencing data available showed that mRNA levels of NTRK3 (hazard ratio [HR] = 0.53, p = 0.021), IGF1R (HR = 1.82, p = 0.027) and KDR (HR = 0.50, p = 0.011) were independently associated with progression-free survival (PFS). Based on the expression of these multi-receptor tyrosine kinase genes, i.e. the features NTRK3-high, IGF1R-low and KDR-high, we developed a pazopanib efficacy predictor that stratified patients into three groups with significantly different PFS (p < 0.0001). Application of the pazopanib efficacy predictor to an independent cohort of patients with pazopanib-treated sarcoma from DKTK MASTER (n = 43) confirmed its potential to separate patient groups with significantly different PFS (p = 0.02), whereas no such association was observed in patients with sarcoma from DKTK MASTER (n = 97) or The Cancer Genome Atlas sarcoma cohort (n = 256) who were not treated with pazopanib. CONCLUSION A score based on the combined expression of NTRK3, IGF1R and KDR allows the identification of patients with sarcoma and with good, intermediate and poor outcome following pazopanib therapy and warrants prospective investigation as a predictive tool to optimise the use of this drug in the clinic.
Collapse
Affiliation(s)
- Christoph E Heilig
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany. https://twitter.com/ChrisHeiligMD
| | - Andreas Laßmann
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sadaf S Mughal
- Division of Applied Bioinformatics, DKFZ, Heidelberg, Germany
| | - Andreas Mock
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; Department of Medical Oncology, NCT Heidelberg and Heidelberg University Hospital, Heidelberg, Germany. https://twitter.com/am0ck
| | - Sebastian Pirmann
- Computational Oncology Group, Molecular Precision Oncology Program, NCT Heidelberg and DKFZ, Heidelberg, Germany
| | - Veronica Teleanu
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marcus Renner
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Carolin Andresen
- Pattern Recognition and Digital Medicine Group, Heidelberg Institute for Stem Cell Technology and Experimental Medicine, Heidelberg, Germany
| | - Bruno C Köhler
- Department of Medical Oncology, NCT Heidelberg and Heidelberg University Hospital, Heidelberg, Germany. https://twitter.com/koehlerlab
| | - Bogac Aybey
- Division of Applied Bioinformatics, DKFZ, Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Sebastian Bauer
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany; DKTK, Essen, Germany. https://twitter.com/seppobauer
| | - Jens T Siveke
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany; DKTK, Essen, Germany; Division of Solid Tumor Translational Oncology, DKTK, Essen, and DKFZ, Heidelberg, Germany; Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Rainer Hamacher
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany; DKTK, Essen, Germany
| | - Gunnar Folprecht
- Department of Medicine I, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Stephan Richter
- Department of Medicine I, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Evelin Schröck
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany; Center for Personalized Oncology, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany; DKTK, Dresden, Germany
| | - Christian H Brandts
- University Cancer Center (UCT) Frankfurt, University Hospital Frankfurt, Goethe University, Frankfurt, Germany; Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany; Frankfurt Cancer Institute, Frankfurt, Germany; DKTK, Frankfurt, Germany
| | - Marit Ahrens
- Department of Medicine, Hematology/Oncology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Peter Hohenberger
- Department of Surgery, Mannheim University Medical Center, Heidelberg University, Mannheim, Germany; Sarcoma Unit, Interdisciplinary Tumor Center Mannheim, Mannheim University Medical Center, Heidelberg University, Mannheim, Germany
| | - Gerlinde Egerer
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Kindler
- UCT Mainz, Johannes Gutenberg University Mainz, Mainz, Germany; Department of Hematology, Medical Oncology and Pneumology, University Medical Center, Mainz, Germany; DKTK, Mainz, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, University of Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Comprehensive Cancer Center Freiburg, University of Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany; DKTK, Freiburg, Germany
| | - Anna L Illert
- Comprehensive Cancer Center Freiburg, University of Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany; DKTK, Freiburg, Germany; Department of Internal Medicine I, University of Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nikolas von Bubnoff
- Department of Internal Medicine I, University of Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Lübeck, Germany
| | - Leonidas Apostolidis
- Department of Medical Oncology, NCT Heidelberg and Heidelberg University Hospital, Heidelberg, Germany
| | - Philipp J Jost
- Department of Hematology and Oncology, Klinikum Rechts der Isar, Technical University Munich, Munich, Germany; Division of Clinical Oncology, Department of Medicine, Medical University of Graz, Graz, Austria; DKTK, Munich, Germany
| | - C Benedikt Westphalen
- DKTK, Munich, Germany; Comprehensive Cancer Center, University Hospital, Ludwig Maximilians University Munich, Munich, Germany; Department of Medicine III, University Hospital, Ludwig Maximilians University Munich, Munich, Germany
| | - Wilko Weichert
- DKTK, Munich, Germany; Institute of Pathology, Technical University Munich, Munich, Germany
| | - Ulrich Keilholz
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Berlin, Germany; DKTK, Berlin, Germany
| | - Frederick Klauschen
- DKTK, Berlin, Germany; Institute of Pathology, Charité - Universitätsmedizin Berlin, And Berlin Institute of Health, Berlin, Germany; Institute of Pathology, Ludwig Maximilians University Munich, Munich, Germany
| | - Katja Beck
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Ulrike Winter
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Daniela Richter
- Center for Personalized Oncology, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany; DKTK, Dresden, Germany; Department of Translational Medical Oncology, NCT Dresden and DKFZ, Dresden, Germany
| | - Lino Möhrmann
- Center for Personalized Oncology, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany; DKTK, Dresden, Germany; Department of Translational Medical Oncology, NCT Dresden and DKFZ, Dresden, Germany
| | - Michael Bitzer
- Department of Internal Medicine I, University Hospital, Eberhard-Karls University, Tübingen, Germany; DKTK, Tübingen, Germany
| | - Klaus Schulze-Osthoff
- DKTK, Tübingen, Germany; Department of Molecular Medicine, Interfaculty Institute for Biochemistry, University of Tübingen, Tübingen, Germany
| | - Benedikt Brors
- German Cancer Consortium (DKTK), Heidelberg, Germany; Division of Applied Bioinformatics, DKFZ, Heidelberg, Germany
| | | | - Simon Kreutzfeldt
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Christoph Heining
- Center for Personalized Oncology, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany; DKTK, Dresden, Germany; Department of Translational Medical Oncology, NCT Dresden and DKFZ, Dresden, Germany. https://twitter.com/ChrisHeining
| | - Daniel B Lipka
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany. https://twitter.com/dblipka1
| | - Albrecht Stenzinger
- German Cancer Consortium (DKTK), Heidelberg, Germany; Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Richard F Schlenk
- German Cancer Consortium (DKTK), Heidelberg, Germany; Department of Medical Oncology, NCT Heidelberg and Heidelberg University Hospital, Heidelberg, Germany; Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany; NCT Trial Center, NCT Heidelberg and DKFZ, Heidelberg, Germany
| | - Peter Horak
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany. https://twitter.com/PeterHorak_MD
| | - Hanno Glimm
- Center for Personalized Oncology, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany; DKTK, Dresden, Germany; Department of Translational Medical Oncology, NCT Dresden and DKFZ, Dresden, Germany
| | - Daniel Hübschmann
- German Cancer Consortium (DKTK), Heidelberg, Germany; Computational Oncology Group, Molecular Precision Oncology Program, NCT Heidelberg and DKFZ, Heidelberg, Germany; Pattern Recognition and Digital Medicine Group, Heidelberg Institute for Stem Cell Technology and Experimental Medicine, Heidelberg, Germany
| | - Stefan Fröhling
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
13
|
Liu S, Yu J, Zhang H, Liu J. TP53 Co-Mutations in Advanced EGFR-Mutated Non-Small Cell Lung Cancer: Prognosis and Therapeutic Strategy for Cancer Therapy. Front Oncol 2022; 12:860563. [PMID: 35444951 PMCID: PMC9013831 DOI: 10.3389/fonc.2022.860563] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/16/2022] [Indexed: 12/25/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. As the most prevalent molecular mutation subtypes in non-small cell lung cancer (NSCLC), EGFR-TKIs are currently a standard first-line therapy for targeting the mutated EGFR in advanced NSCLC patients. However, 20-30% of this subset of patients shows primary resistance to EGFR-TKIs. Patients with co-mutations of EGFR and several other genes have a poor response to EGFR-TKIs, whereas the prognostic and predictive significance of EGFR/TP53 co-mutation in NSCLC patients remains controversial. Meanwhile, little is known about how to choose an optimal therapeutic strategy for this subset of patients. Presently, no drugs targeting TP53 mutations are available on the market, and some p53 protein activators are in the early stage of clinical trials. A combination of EGFR-TKIs with antiangiogenic agents or chemotherapy or other agents might be a more appropriate strategy to tackle the problem. In this review, we describe the prognostic and predictive value of EGFR/TP53 co-mutation in NSCLC patients, investigate the mechanisms of this co-mutation affecting the response to EGFR-TKIs, and further explore optimal regimens effectively to prolong the survival time of the NSCLC patients harboring this co-mutation.
Collapse
Affiliation(s)
- Surui Liu
- Department of Oncology, Jinan Central Hospital, Jinan, China.,Department of Oncology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Jin Yu
- Department of Oncology, Jinan Central Hospital, Jinan, China
| | - Hui Zhang
- Department of Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jie Liu
- Department of Oncology, Jinan Central Hospital, Jinan, China
| |
Collapse
|
14
|
Elisabetta G, Anna B, Adriano P, Andrea CD, Guido S, Ilaria P, Andrea B, Lorenzo A, Serena P. Pharmacogenomics of soft tissue sarcomas: New horizons to understand efficacy and toxicity. Cancer Treat Res Commun 2022; 31:100528. [PMID: 35123198 DOI: 10.1016/j.ctarc.2022.100528] [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: 10/13/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 01/27/2023]
Abstract
Clinical responses to anticancer therapies in advanced soft tissue sarcoma (STS) are unfortunately limited to a small subset of patients. Much of the inter-individual variability in treatment efficacy and risk of toxicities is as result of polymorphisms in genes encoding proteins involved in drug pharmacokinetics and pharmacodynamics. Therefore, the detection of pharmacogenomics (PGx) biomarkers that might predict drug response and toxicity can be useful to explain the genetic basis for the differences in treatment efficacy and toxicity among STS patients. PGx markers are frequently located in transporters, drug-metabolizing enzyme genes, drug targets, or HLA alleles. Along this line, genetic variability harbouring in the germline genome of the patients can influence systemic pharmacokinetics and pharmacodynamics of the treatments, acting as predictive biomarkers for drug-induced toxicity and treatment efficacy. By linking drug activity to the functional complexity of cancer genomes, also systematic pharmacogenomic profiling in cancer cell lines and primary STS samples represents area of active investigation that could eventually lead to enhanced efficacy and offer a powerful biomarker discovery platform to optimize current treatments and improve the knowledge about the individual's drug response in STS patients into the clinical practice.
Collapse
Affiliation(s)
| | - Boddi Anna
- Department of Health Science, University of Florence, Florence, Italy
| | - Pasqui Adriano
- Medical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Campanacci Domenico Andrea
- Department of Health Science, University of Florence, Florence, Italy; Department of Orthopaedic Oncology and Reconstructive Surgery, Careggi University Hospital, Florence, Italy
| | - Scoccianti Guido
- Department of Health Science, University of Florence, Florence, Italy
| | - Palchetti Ilaria
- Department of Chemistry Ugo Schiff, University of Florence, Sesto Fiorentino, Florence, Italy
| | - Bernini Andrea
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, Siena, 53100 Italy
| | - Antonuzzo Lorenzo
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy; Medical Oncology Unit, Careggi University Hospital, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Pillozzi Serena
- Medical Oncology Unit, Careggi University Hospital, Florence, Italy.
| |
Collapse
|
15
|
Multiparametric Circulating Tumor Cell Analysis to Select Targeted Therapies for Breast Cancer Patients. Cancers (Basel) 2021; 13:cancers13236004. [PMID: 34885114 PMCID: PMC8657376 DOI: 10.3390/cancers13236004] [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: 10/22/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Liquid biopsies may act as a dynamic tool for identification of targets for precision therapy while circumventing limitations of tissue biopsies. In opposite to most liquid biopsy-related studies that analyze limited patient material for only one parameter, this study is based on a longitudinal and multiparametric analysis of circulating tumor cells (CTCs). A metastatic breast cancer patient was followed over a period of three years and analyses of the genome, RNA profiling, and in vitro drug testing on cultured CTCs were performed in a unique manner. We show that combining the strengths of multiple technologies for analysis yielded maximum information on the ongoing disease and, eventually, allowed choosing an effective therapy, which led to a massive reduction in CTC numbers. This approach provides a concept for future detailed longitudinal and multiparametric CTC analyses. Abstract Background: The analysis of liquid biopsies, e.g., circulating tumor cells (CTCs) is an appealing diagnostic concept for targeted therapy selection. In this proof-of-concept study, we aimed to perform multiparametric analyses of CTCs to select targeted therapies for metastatic breast cancer patients. Methods: First, CTCs of five metastatic breast cancer patients were analyzed by whole exome sequencing (WES). Based on the results, one patient was selected and monitored by longitudinal and multiparametric liquid biopsy analyses over more than three years, including WES, RNA profiling, and in vitro drug testing of CTCs. Results: Mutations addressable by targeted therapies were detected in all patients, including mutations that were not detected in biopsies of the primary tumor. For the index patient, the clonal evolution of the tumor cells was retraced and resistance mechanisms were identified. The AKT1 E17K mutation was uncovered as the driver of the metastatic process. Drug testing on the patient’s CTCs confirmed the efficacy of drugs targeting the AKT1 pathway. During a targeted therapy chosen based on the CTC characterization and including the mTOR inhibitor everolimus, CTC numbers dropped by 97.3% and the disease remained stable as determined by computer tomography/magnetic resonance imaging. Conclusion: These results illustrate the strength of a multiparametric CTC analysis to choose and validate targeted therapies to optimize cancer treatment in the future. Furthermore, from a scientific point of view, such studies promote the understanding of the biology of CTCs during different treatment regimens.
Collapse
|
16
|
Noor H, Briggs NE, McDonald KL, Holst J, Vittorio O. TP53 Mutation Is a Prognostic Factor in Lower Grade Glioma and May Influence Chemotherapy Efficacy. Cancers (Basel) 2021; 13:5362. [PMID: 34771529 PMCID: PMC8582451 DOI: 10.3390/cancers13215362] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/17/2021] [Accepted: 10/22/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Identification of prognostic biomarkers in cancers is a crucial step to improve overall survival (OS). Although mutations in tumour protein 53 (TP53) is prevalent in astrocytoma, the prognostic effects of TP53 mutation are unclear. METHODS In this retrospective study, we sequenced TP53 exons 1 to 10 in a cohort of 102 lower-grade glioma (LGG) subtypes and determined the prognostic effects of TP53 mutation in astrocytoma and oligodendroglioma. Publicly available datasets were analysed to confirm the findings. RESULTS In astrocytoma, mutations in TP53 codon 273 were associated with a significantly increased OS compared to the TP53 wild-type (HR (95% CI): 0.169 (0.036-0.766), p = 0.021). Public datasets confirmed these findings. TP53 codon 273 mutant astrocytomas were significantly more chemosensitive than TP53 wild-type astrocytomas (HR (95% CI): 0.344 (0.13-0.88), p = 0.0148). Post-chemotherapy, a significant correlation between TP53 and YAP1 mRNA was found (p = 0.01). In O (6)-methylguanine methyltransferase (MGMT) unmethylated chemotherapy-treated astrocytoma, both TP53 codon 273 and YAP1 mRNA were significant prognostic markers. In oligodendroglioma, TP53 mutations were associated with significantly decreased OS. CONCLUSIONS Based on these findings, we propose that certain TP53 mutant astrocytomas are chemosensitive through the involvement of YAP1, and we outline a potential mechanism. Thus, TP53 mutations may be key drivers of astrocytoma therapeutic efficacy and influence survival outcomes.
Collapse
Affiliation(s)
- Humaira Noor
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, University of New South Wales, Sydney, NSW 2031, Australia;
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Randwick, NSW 2031, Australia;
| | - Nancy E. Briggs
- Stats Central, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW 2031, Australia;
| | - Kerrie L. McDonald
- Cure Brain Cancer Biomarkers and Translational Research Group, Prince of Wales Clinical School, University of New South Wales, Sydney, NSW 2031, Australia;
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Randwick, NSW 2031, Australia;
| | - Jeff Holst
- Adult Cancer Program, Lowy Cancer Research Centre, UNSW Sydney, Randwick, NSW 2031, Australia;
- Translational Cancer Metabolism Laboratory, School of Medical Sciences, Prince of Wales Clinical School, UNSW Sydney, Sydney, NSW 2031, Australia
| | - Orazio Vittorio
- School of Women’s & Children’s Health, UNSW Medicine, University of NSW, Randwick, NSW 2031, Australia;
- Children’s Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Randwick, NSW 2031, Australia
| |
Collapse
|
17
|
Sicklick JK, Kato S, Okamura R, Patel H, Nikanjam M, Fanta PT, Hahn ME, De P, Williams C, Guido J, Solomon BM, McKay RR, Krie A, Boles SG, Ross JS, Lee JJ, Leyland-Jones B, Lippman SM, Kurzrock R. Molecular profiling of advanced malignancies guides first-line N-of-1 treatments in the I-PREDICT treatment-naïve study. Genome Med 2021; 13:155. [PMID: 34607609 PMCID: PMC8491393 DOI: 10.1186/s13073-021-00969-w] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 09/15/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Malignancies are molecularly complex and become more resistant with each line of therapy. We hypothesized that offering matched, individualized combination therapies to patients with treatment-naïve, advanced cancers would be feasible and efficacious. Patients with newly diagnosed unresectable/metastatic, poor-prognosis cancers were enrolled in a cross-institutional prospective study. METHODS A total of 145 patients were included in the study. Genomic profiling (tissue and/or circulating tumor DNA) was performed in all patients, and PD-L1 immunohistochemistry, tumor mutational burden, and microsatellite status assessment were performed in a subset of patients. We evaluated safety and outcomes: disease-control rate (stable disease for ≥ 6 months or partial or complete response), progression-free survival (PFS), and overall survival (OS). RESULTS Seventy-six of 145 patients (52%) were treated, most commonly for non-colorectal gastrointestinal cancers, carcinomas of unknown primary, and hepatobiliary malignancies (53% women; median age, 63 years). The median number of deleterious genomic alterations per patient was 5 (range, 0-15). Fifty-four treated patients (71%) received ≥ 1 molecularly matched therapy, demonstrating the feasibility of administering molecularly matched therapy. The Matching Score, which reflects the percentage of targeted alterations, correlated linearly with progression-free survival (R2 = 0.92; P = 0.01), and high (≥ 60%) Matching Score was an independent predictor of improved disease control rate [OR 3.31 (95% CI 1.01-10.83), P = 0.048], PFS [HR 0.55 (0.28-1.07), P = 0.08], and OS [HR 0.42 (0.21-0.85), P = 0.02]. Serious adverse event rates were similar in the unmatched and matched groups. CONCLUSIONS Personalized combination therapies targeting a majority of a patient's molecular alterations have antitumor activity as first-line treatment. These findings underscore the feasibility and importance of using tailored N-of-1 combination therapies early in the course of lethal malignancies. TRIAL REGISTRATION I-PREDICT ( NCT02534675 ) was registered on August 25, 2015.
Collapse
Affiliation(s)
- Jason K Sicklick
- Department of Surgery, Division of Surgical Oncology, UC San Diego School of Medicine, San Diego, CA, USA.
- Center for Personalized Cancer Therapy, Moores Cancer Center, UC San Diego Health, 3855 Health Sciences Drive, Mail Code 0658, La Jolla, CA, 92093-0658, USA.
| | - Shumei Kato
- Center for Personalized Cancer Therapy, Moores Cancer Center, UC San Diego Health, 3855 Health Sciences Drive, Mail Code 0658, La Jolla, CA, 92093-0658, USA
- Department of Medicine, Division of Hematology Oncology, UC San Diego School of Medicine, San Diego, CA, USA
| | - Ryosuke Okamura
- Center for Personalized Cancer Therapy, Moores Cancer Center, UC San Diego Health, 3855 Health Sciences Drive, Mail Code 0658, La Jolla, CA, 92093-0658, USA
- Department of Medicine, Division of Hematology Oncology, UC San Diego School of Medicine, San Diego, CA, USA
| | - Hitendra Patel
- Center for Personalized Cancer Therapy, Moores Cancer Center, UC San Diego Health, 3855 Health Sciences Drive, Mail Code 0658, La Jolla, CA, 92093-0658, USA
- Department of Medicine, Division of Hematology Oncology, UC San Diego School of Medicine, San Diego, CA, USA
| | - Mina Nikanjam
- Center for Personalized Cancer Therapy, Moores Cancer Center, UC San Diego Health, 3855 Health Sciences Drive, Mail Code 0658, La Jolla, CA, 92093-0658, USA
- Department of Medicine, Division of Hematology Oncology, UC San Diego School of Medicine, San Diego, CA, USA
| | - Paul T Fanta
- Center for Personalized Cancer Therapy, Moores Cancer Center, UC San Diego Health, 3855 Health Sciences Drive, Mail Code 0658, La Jolla, CA, 92093-0658, USA
- Department of Medicine, Division of Hematology Oncology, UC San Diego School of Medicine, San Diego, CA, USA
| | - Michael E Hahn
- Department of Radiology, UC San Diego School of Medicine, San Diego, CA, USA
| | - Pradip De
- Avera Cancer Institute, Sioux Falls, SD, USA
| | | | - Jessica Guido
- Center for Personalized Cancer Therapy, Moores Cancer Center, UC San Diego Health, 3855 Health Sciences Drive, Mail Code 0658, La Jolla, CA, 92093-0658, USA
| | | | - Rana R McKay
- Center for Personalized Cancer Therapy, Moores Cancer Center, UC San Diego Health, 3855 Health Sciences Drive, Mail Code 0658, La Jolla, CA, 92093-0658, USA
- Department of Medicine, Division of Hematology Oncology, UC San Diego School of Medicine, San Diego, CA, USA
| | - Amy Krie
- Avera Cancer Institute, Sioux Falls, SD, USA
| | - Sarah G Boles
- Center for Personalized Cancer Therapy, Moores Cancer Center, UC San Diego Health, 3855 Health Sciences Drive, Mail Code 0658, La Jolla, CA, 92093-0658, USA
- Department of Medicine, Division of Hematology Oncology, UC San Diego School of Medicine, San Diego, CA, USA
| | - Jeffrey S Ross
- Foundation Medicine, Inc., Cambridge, MA, USA
- Departments of Pathology and Urology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - J Jack Lee
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Scott M Lippman
- Center for Personalized Cancer Therapy, Moores Cancer Center, UC San Diego Health, 3855 Health Sciences Drive, Mail Code 0658, La Jolla, CA, 92093-0658, USA
- Department of Medicine, Division of Hematology Oncology, UC San Diego School of Medicine, San Diego, CA, USA
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy, Moores Cancer Center, UC San Diego Health, 3855 Health Sciences Drive, Mail Code 0658, La Jolla, CA, 92093-0658, USA.
- Department of Medicine, Division of Hematology Oncology, UC San Diego School of Medicine, San Diego, CA, USA.
| |
Collapse
|
18
|
Zou J, Yang X, Duan J, Wang J, Yang Z, Luo D, Liu L, Chen J, Nie J. A Case Report of Targeted Therapy with Anlotinib in a Patient with Advanced Breast Metaplastic Carcinoma. Onco Targets Ther 2021; 14:4599-4607. [PMID: 34475766 PMCID: PMC8408045 DOI: 10.2147/ott.s318645] [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: 05/11/2021] [Accepted: 08/11/2021] [Indexed: 12/13/2022] Open
Abstract
Metaplastic breast carcinoma (MBC) is a rare malignant breast tumor, and no effective chemotherapy unique to metaplastic carcinoma exists. As MBC is typically “triple negative”, endocrine therapy and molecular therapy targeted to Her2 might not be favorable, resulting in a poor prognosis. Anlotinib is currently being tested in patients with breast or cancer. Here, we report a successful case in which anlotinib was used to treat MBC. A 54-year-old female patient visited the hospital after the discovery of a left breast tumor 10 months prior, and tumor redness and swelling had lasted for more than one month. After admission, relevant examinations were performed. After left breast tumor puncture revealed left emulsified biological cancer, the tumor significantly increased in size, and bleeding was obvious after 2 cycles of the “EC” chemotherapy regimen. The curative effect was evaluated as progressive disease (PD). After two cycles of chemotherapy with the “PCb” regimen, the efficacy was still PD. The Karnofsky performance status (KPS) score of the patient after 4 cycles of chemotherapy was 60 points, with severe anemia, and she could not tolerate chemotherapy. The patient was given radiotherapy to stop bleeding, and the tumor further increased in size during radiotherapy. The curative effect was evaluated as PD. After a multidisciplinary consultation in our hospital, we initiated oral anlotinib (12 mg; 2 weeks on, 1 week off). The tumor significantly decreased in size after taking anlotinib, and the efficacy was evaluated as PR. Adverse reactions during treatment were controlled, and progression-free survival (PFS) reached up to 25+ months. The follow-up is ongoing. The patient has provided written informed consent for the case details and images to be published, and at the same time institutional approval was required to publish the case details, we report this case.
Collapse
Affiliation(s)
- Jieya Zou
- The Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, People's Republic of China
| | - Xiaojuan Yang
- The Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, People's Republic of China
| | - Jiajun Duan
- The Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, People's Republic of China
| | - Ji Wang
- The Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, People's Republic of China
| | - Zhuangqing Yang
- The Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, People's Republic of China
| | - Dan Luo
- Department of Pathology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, People's Republic of China
| | - Lihua Liu
- The Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, People's Republic of China
| | - Junyao Chen
- The Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, People's Republic of China
| | - Jianyun Nie
- The Third Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, People's Republic of China
| |
Collapse
|
19
|
Genomic landscape and tumor mutation burden analysis of Chinese patients with sarcomatoid carcinoma of the head and neck. Oral Oncol 2021; 121:105436. [PMID: 34371452 DOI: 10.1016/j.oraloncology.2021.105436] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 01/23/2023]
Abstract
BACKGROUND Sarcomatoid carcinoma (SC) of the head and neck (HN) is a rare disease that has both sarcomatoid and cancerous components. The genetic background and mechanisms of tumorigenesis remain largely unrevealed, and the progress of precision therapy has been limited. METHODS Targeted DNA-based next-generation sequencing (NGS) was performed by a 539 genes panel of pan-cancer in 12 patients with SC of the HN to identify their genetic alterations and investigate clinically actionable mutations for use in precision treatment. RESULTS TP53 was identified as the most frequently mutated gene. Genes related to the cell cycling, chromatin remodeling and histone modification were found to be frequently mutated in patients with SC of the HN. Alterations in receptor tyrosine kinases (RTKs) were also found in six patients. In addition, four patients had mutations in members of the downstream RAS and PI3-kinase pathways, PIK3CA was identified as the most frequently mutated gene in this pathway. The tumor mutation burden (TMB) value ranged from 0.71 to 14.71 per megabase, with a median of 4.34. The TMB value of PIK3CA mutation patients was significantly higher than that of PIK3CA wild-type patients. CONCLUSIONS This was the first study to investigate genomic alterations specifically in Chinese patients with SC of the HN. Our research results showed that 10 out of 12 patients can match the targeted therapies or immunotherapy currently available in clinical practice or active clinical trials, suggesting precision therapy has the potential utility to improve the long-term prognosis for patients with the rare disease. Due to the small number of patients in this study, the findings need to be validated in a larger cohort.
Collapse
|
20
|
Hao J, Peng Q, Wang K, Yu G, Pan Y, Du X, Hu N, Zhang X, Qin Y, Li H. Antitumor Effect of Lenvatinib Combined with Alisertib in Hepatocellular Carcinoma by Targeting the DNA Damage Pathway. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6613439. [PMID: 34337035 PMCID: PMC8324353 DOI: 10.1155/2021/6613439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 06/12/2021] [Accepted: 07/09/2021] [Indexed: 12/24/2022]
Abstract
METHODS Immunohistochemical staining, sequencing, and genetic analysis of liver cancer tissues were performed. The antitumor efficacy of single-agent or combination treatment was measured by cell counting kit-8 assay and colony formation assays. Their antiproliferative and apoptosis activity is evaluated by cell cycle analyses and wound healing assays. The DNA-related proteins were also measured by Western blotting and immunohistochemical staining. The HepG2 xenograft model was used to detect the effects of lenvatinib-alisertib on the antitumor activity. RESULTS AURKA was found to be upregulated in HCC tissues (77.3%, 17/22). Combined alisertib and lenvatinib treatment significantly enhanced the inhibition of proliferation and migration in HepG2 and Hep3B cell lines compared to single-agent treatments (all Ps < 0.01). Alisertib alone or in combination with lenvatinib demonstrated a significant increase in the percentage of super-G2 cells (lenvatinib 1 μM vs. lenvatinib 1 μM + alisertib 0.1 μM 8.84 ± 0.84 vs. 34.0 ± 1.54, P < 0.001). Discontinuous spindles and missegregated chromosomes in HCC cells treated with alisertib in combination with lenvatinib were observed. We further revealed that combined treatment inhibited the expression of DNA damage pathway proteins compared to those of single-agent treatments. In nude mice, combined administration of alisertib combined with lenvatinib significantly enhanced the suppression of tumor growth and induced apoptosis (all Ps < 0.01). CONCLUSIONS Our findings provide evidence for the possible use of alisertib in combination with lenvatinib in the treatment of HCC for better therapeutic outcomes.
Collapse
Affiliation(s)
- Jianwen Hao
- Department of Radiology, Tianjin Chest Hospital, Tianjin 300350, China
| | - Qizhen Peng
- Department of Radiology, Tungwah Hospital of Sun Yat-Sen University, Dongguan, 523000 Guangdong, China
| | - Keruo Wang
- Department of Diagnostics, Tianjin Medical University, Tianjin 300070, China
| | - Ge Yu
- Department of Hepatobiliary, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Clinical Research Center for Cancer, Tianjin 300070, China
| | - Yi Pan
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Clinical Research Center for Cancer, Tianjin 300070, China
| | - Xiaoling Du
- Department of Diagnostics, Tianjin Medical University, Tianjin 300070, China
| | - Na Hu
- Department of Diagnostics, Tianjin Medical University, Tianjin 300070, China
| | - Xuening Zhang
- Department of Radiology Second Hospital of Tianjin Medical University, Tianjin 300070, China
| | - Yu Qin
- Department of Diagnostics, Tianjin Medical University, Tianjin 300070, China
| | - Huikai Li
- Department of Hepatobiliary, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Clinical Research Center for Cancer, Tianjin 300070, China
| |
Collapse
|
21
|
Gaiser T, Sauer C, Marx A, Jakob J, Kasper B, Hohenberger P, Hirsch D, Ronellenfitsch U. Molecular and Pathological Profiling of Corresponding Treatment-Naïve and Neoadjuvant Pazopanib-Treated High-Risk Soft Tissue Sarcoma Samples of the GISG-04/NOPASS Study. BIOLOGY 2021; 10:biology10070639. [PMID: 34356494 PMCID: PMC8301157 DOI: 10.3390/biology10070639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/07/2021] [Indexed: 01/25/2023]
Abstract
In the framework of the German Interdisciplinary Sarcoma Group GISG-04/NOPASS trial, we evaluated soft tissue sarcoma samples taken before and after neoadjuvant pazopanib therapy using histopathology and next generation sequencing (NGS) to find potential predictive biomarkers. We also aimed to improve the genetically based sarcoma classification and to elucidate additional potentially druggable mutations. In total, 30 tumor samples from 18 patients consisting of 12 pre-therapeutic biopsies and 18 resection specimens following neoadjuvant pazopanib therapy were available for analyses. NGS was performed with the Oncomine Focus Assay (Ion Torrent) covering 0.03 Mb of DNA and enabled the detection of genetic variants in 52 cancer-relevant genes. Pathological analysis showed significant regression (≥50%) after pazopanib treatment in only one undifferentiated (pleomorphic) sarcoma. NGS analyses revealed a very high frequency of CDK4 amplification (88%; 7/8) in the group of dedifferentiated liposarcoma. In addition, two potentially druggable mutations, a MAP2K1 missense mutation (E203K) and a BRAF missense mutation (V600E), were traceable in two undifferentiated (pleomorphic) sarcoma patients (11%; 2/18). Our findings demonstrate that NGS testing is a powerful technology helping to improve diagnostic accuracy and offering some patients the chance for personalized medicine even in a "mutation unlikely" cohort like STS.
Collapse
Affiliation(s)
- Timo Gaiser
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, 68305 Mannheim, Germany; (C.S.); (A.M.); (D.H.)
- Correspondence: ; Tel.: +49-621-383-2876; Fax: +49-621-383-2005
| | - Christian Sauer
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, 68305 Mannheim, Germany; (C.S.); (A.M.); (D.H.)
| | - Alexander Marx
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, 68305 Mannheim, Germany; (C.S.); (A.M.); (D.H.)
| | - Jens Jakob
- Department of General, Visceral and Child Surgery, University Medical Center Göttingen, 37075 Göttingen, Germany;
| | - Bernd Kasper
- Sarcoma Unit, Interdisciplinary Tumor Center Mannheim, Mannheim University Medical Center, 68305 Mannheim, Germany;
| | - Peter Hohenberger
- Division of Surgical Oncology and Thoracic Surgery, University Medical Center Mannheim, 68167 Mannheim, Germany;
| | - Daniela Hirsch
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, 68305 Mannheim, Germany; (C.S.); (A.M.); (D.H.)
| | - Ulrich Ronellenfitsch
- Department of Abdominal, Vascular, and Endocrine Surgery, University Hospital Halle (Saale), 06120 Halle (Saale), Germany;
| |
Collapse
|
22
|
Nassif EF, Auclin E, Bahleda R, Honoré C, Mir O, Dumont S, Mery B, Hodroj K, Brahmi M, Trédan O, Ray-Coquard I, Blay JY, Massard C, Le Cesne A, Dufresne A. TP53 Mutation as a Prognostic and Predictive Marker in Sarcoma: Pooled Analysis of MOSCATO and ProfiLER Precision Medicine Trials. Cancers (Basel) 2021; 13:3362. [PMID: 34282771 PMCID: PMC8268242 DOI: 10.3390/cancers13133362] [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: 06/08/2021] [Revised: 06/23/2021] [Accepted: 07/02/2021] [Indexed: 11/16/2022] Open
Abstract
(1) Background: locally resected high-grade sarcomas relapse in 40% of cases. There is no prognostic or predictive genomic marker for response to peri-operative chemotherapy. (2) Methods: MOSCATO and ProfiLER are pan-tumor prospective precision medicine trials for advanced tumors. Molecular analysis in both trials comprised targeted next-generation sequencing and comparative genomic hybridization array. We investigated if molecular alterations identified in these trials in sarcomas were associated with disease-free survival (DFS) and response to anthracyclines. (3) Results: this analysis included 215 sarcomas, amongst which 53 leiomyosarcomas, 27 rhabdomyosarcomas, 20 undifferentiated pleomorphic sarcomas, and 17 liposarcomas. The most frequently altered gene was TP53 (46 mutations and eight deletions). There were 149 surgically resected localized sarcomas. Median DFS in TP53 wild type (WT), deleted, and mutated sarcomas was 16, 10, and 10 months, respectively (p = 0.028; deletions: HR = 1.55; 95% CI = 0.75-3.19; mutations: HR = 1.70; 95%CI = 1.13-2.64). In multivariate analysis, TP53 mutations remained associated with shorter DFS (p = 0.027; HR = 2.30; 95%CI = 1.10-4.82). There were 161 localized and advanced sarcomas evaluable for response to anthracyclines. Objective response rates were 35% and 55% in TP53 WT and mutated sarcomas, respectively (OR = 2.24; 95%CI = 1.01-5.03; p = 0.05). In multivariate analysis, TP53 mutations remained associated with increased response (OR = 3.24; 95%CI = 1.30-8.45; p = 0.01). (4) Conclusions: TP53 mutations are associated with shorter DFS and increased response to anthracyclines. Post-validation, these findings could assist in decision-making for peri-operative treatments.
Collapse
Affiliation(s)
- Elise F. Nassif
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| | - Edouard Auclin
- Oncology Department, Hopital Européen Georges Pompidou, 75015 Paris, France;
| | - Rastilav Bahleda
- DITEP (Département d’Innovation Therapeutique et d’Essais Précoces), Drug Development Department, Gustave Roussy, 94805 Villejuif, France; (R.B.); (C.M.)
| | - Charles Honoré
- Surgical Oncology Department, Gustave Roussy, 94805 Villejuif, France;
| | - Olivier Mir
- Ambulatory Cancer Care Department, Gustave Roussy, 94805 Villejuif, France;
| | - Sarah Dumont
- Medical Oncology Department, Gustave Roussy, 94805 Villejuif, France;
| | - Benoite Mery
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| | - Khalil Hodroj
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| | - Mehdi Brahmi
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| | - Olivier Trédan
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| | - Isabelle Ray-Coquard
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| | - Jean-Yves Blay
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| | - Christophe Massard
- DITEP (Département d’Innovation Therapeutique et d’Essais Précoces), Drug Development Department, Gustave Roussy, 94805 Villejuif, France; (R.B.); (C.M.)
| | - Axel Le Cesne
- Medical Oncology Department, Gustave Roussy, 94805 Villejuif, France;
| | - Armelle Dufresne
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| |
Collapse
|
23
|
Akrivi S, Varras M, Anastasiadi Z, Pappa C, Vlachioti A, Varra VK, Varra FN, Balasi E, Akrivis C. Primary vulvar leiomyosarcoma localized in the Bartholin's gland area: A case report and review. Mol Clin Oncol 2021; 14:69. [PMID: 33680460 PMCID: PMC7890440 DOI: 10.3892/mco.2021.2231] [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: 03/13/2020] [Accepted: 01/14/2021] [Indexed: 01/14/2023] Open
Abstract
Vulvar sarcomas located in the Bartholin's gland area are extremely uncommon mesenchymal vulvar tumors. These neoplasms can be mistaken as Bartholin' gland benign lesions such as cysts or abscesses, leading to a delay in the diagnosis of underlying malignancy. Currently, only a few cases of these aggressive cancers have been reported in the literature. A 42-year-old female patient without any previous complaint presented to Obstetrics and Gynecology Department of ‘G. Chaztikosta’ General Hospital due to a vulvar lump in the area of the left Bartholin's gland with a 6-month history of progressive swelling. Pelvic examination showed a solid mass of 6.5-cm in maximum diameter, localized in the left Bartholin's gland. The patient underwent wide local excision and histopathological examination of hematoxylin and eosin-stained sections indicated intersecting fascicles of spindle cells, with moderate to severe atypia. The number of mitoses was up to 8 per 10 high power fields. The neoplasm to its greatest extent was circumscribed and in places had an invasive growth pattern. Tumoral necrosis was not seen. Involved Bartholin' gland by the tumor was identified. The tumor extended focally to the surgical margin. The neoplastic cells showed positive staining for smooth muscle actin, desmin, HHF35, caldesmon, vimentin and estrogen and progesterone receptors. Immunohistochemistry was negative for S100, myoglobulin, keratin 116, CD117, CD34 and CD31. The patient denied further surgery or/and local radiotherapy, although the mass was >5-cm and a focally infiltrative surgical margin was found. During the close follow-up, no local recurrences or metastases were observed 53 months after surgery. In conclusion, wide local tumor excision with free surgical margins is a good option of surgery for vulvar leiomyosarcomas. In recurrences, a new extensive surgical resection of the lesion and radiotherapy are suggested. Ipsilateral lympadenectomy is indicated when there is a pathologic lymph node. Chemotherapy is provided in cases of distal metastases.
Collapse
Affiliation(s)
- Stella Akrivi
- Department of Obstetrics and Gynecology, Royal Jubille Maternity Hospital, Belfast Trust, Belfast BT12 6BA, UK
| | - Michail Varras
- Fourth Department of Obstetrics and Gynecology, 'Elena Venizelou' General Hospital, Athens 11521, Greece
| | - Zoi Anastasiadi
- Department of Obstetrics and Gynecology, 'G. Chatzikosta' General Hospital, Ioannina 45001, Epirus, Greece
| | - Christina Pappa
- Department of Obstetrics and Gynecology, 'G. Chatzikosta' General Hospital, Ioannina 45001, Epirus, Greece
| | - Aikaterini Vlachioti
- Department of Obstetrics and Gynecology, 'G. Chatzikosta' General Hospital, Ioannina 45001, Epirus, Greece
| | | | - Fani-Niki Varra
- Department of Pharmacy, Frederick University, Nicosia 1036, Cyprus
| | - Eufemia Balasi
- Pathology Department, 'G. Chatzikosta' General Hospital, Ioannina 45001, Epirus, Greece
| | - Christos Akrivis
- Department of Obstetrics and Gynecology, 'G. Chatzikosta' General Hospital, Ioannina 45001, Epirus, Greece
| |
Collapse
|
24
|
Cho WCS, Tse KP, Ngan RKC, Cheuk W, Ma VWS, Yang YT, Yip TTC, Tan KT, Chen SJ. Genomic characterization reveals potential biomarkers in nasopharyngeal carcinoma patients with relapse. Expert Rev Mol Diagn 2020; 20:1149-1159. [PMID: 33040630 DOI: 10.1080/14737159.2020.1835473] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/08/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Although the majority of nasopharyngeal carcinoma (NPC) patients demonstrate favorable outcomes after radiotherapy and/or chemotherapy, about 8-10% of patients will develop recurrent disease, and genomic alterations (GAs) associated with the recurrence are unclear. METHODS This study investigated the GAs in the paired primary tumors and recurrent tumors of 7 NPC patients with relapse, as well as the primary tumors of 15 NPC patients without relapse by deep targeted next-generation sequencing on 440 cancer-related genes. RESULTS BRCA1 and TP53 mutations were significantly enriched in patients with relapse (P = 0.021 and P = 0.023, respectively). Survival analysis revealed that the GAs of TP53, ZNF217, VEGFB, CDKN1B, GNAS, PRDM1, and MEN1 were associated with significantly shorter overall survival. The GAs of the tumor also altered after treatment in the relapsed group, and five genes (CDK4, FGFR3, ALK, BRCA1, and CHEK2) in the recurrent tumors were potentially druggable. CONCLUSIONS The discovery of GAs associated with recurrence or survival in NPC may serve as potential prognostic gene signatures of high-risk patients. Targeted therapies are available in some of the clinically relevant GAs and may be considered in future clinical trials. Given the limitation of the sample size, validation by a larger cohort is warranted.
Collapse
Affiliation(s)
- William C S Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital , Hong Kong SAR, China
| | - Ka-Po Tse
- ACT Genomics, Co. Ltd ., Taipei, Taiwan
| | - Roger K C Ngan
- Department of Clinical Oncology, The University of Hong Kong, Gleneagles Hong Kong Hospital , Hong Kong SAR, China
| | - Wah Cheuk
- Department of Pathology, Queen Elizabeth Hospital , Hong Kong SAR, China
| | - Victor W S Ma
- Department of Clinical Oncology, Queen Elizabeth Hospital , Hong Kong SAR, China
| | | | | | | | | |
Collapse
|
25
|
Kato S, Kim KH, Lim HJ, Boichard A, Nikanjam M, Weihe E, Kuo DJ, Eskander RN, Goodman A, Galanina N, Fanta PT, Schwab RB, Shatsky R, Plaxe SC, Sharabi A, Stites E, Adashek JJ, Okamura R, Lee S, Lippman SM, Sicklick JK, Kurzrock R. Real-world data from a molecular tumor board demonstrates improved outcomes with a precision N-of-One strategy. Nat Commun 2020; 11:4965. [PMID: 33009371 PMCID: PMC7532150 DOI: 10.1038/s41467-020-18613-3] [Citation(s) in RCA: 200] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/25/2020] [Indexed: 01/01/2023] Open
Abstract
Next-generation sequencing (NGS) can identify novel cancer targets. However, interpreting the molecular findings and accessing drugs/clinical trials is challenging. Furthermore, many tumors show resistance to monotherapies. To implement a precision strategy, we initiated a multidisciplinary (basic/translational/clinical investigators, bioinformaticians, geneticists, and physicians from multiple specialties) molecular tumor board (MTB), which included a project manager to facilitate obtaining clinical-grade biomarkers (blood/tissue NGS, specific immunohistochemistry/RNA expression including for immune-biomarkers, per physician discretion) and medication-acquisition specialists/clinical trial coordinators/navigators to assist with medication access. The MTB comprehensively reviewed patient characteristics to develop N-of-One treatments implemented by the treating physician's direction under the auspices of a master protocol. Overall, 265/429 therapy-evaluable patients (62%) were matched to ≥1 recommended drug. Eighty-six patients (20%) matched to all drugs recommended by MTB, including combinatorial approaches, while 38% received physician's choice regimen, generally with unmatched approach/low degree of matching. Our results show that patients who receive MTB-recommended regimens (versus physician choice) have significantly longer progression-free (PFS) and overall survival (OS), and are better matched to therapy. High (≥50%) versus low (<50%) Matching Score therapy (roughly reflecting therapy matched to ≥50% versus <50% of alterations) independently correlates with longer PFS (hazard ratio [HR], 0.63; 95% confidence interval [CI], 0.50-0.80; P < 0.001) and OS (HR, 0.67; 95% CI, 0.50-0.90; P = 0.007) and higher stable disease ≥6 months/partial/complete remission rate (52.1% versus 30.4% P < 0.001) (all multivariate). In conclusion, patients who receive MTB-based therapy are better matched to their genomic alterations, and the degree of matching is an independent predictor of improved oncologic outcomes including survival.
Collapse
Affiliation(s)
- Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA.
| | - Ki Hwan Kim
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA.
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea.
| | - Hyo Jeong Lim
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
- Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Amelie Boichard
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Mina Nikanjam
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Elizabeth Weihe
- Department of Radiology, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Dennis J Kuo
- Division of Pediatric Hematology-Oncology, Rady Children's Hospital-San Diego, University of California San Diego School of Medicine, San Diego, CA, USA
| | - Ramez N Eskander
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Aaron Goodman
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Natalie Galanina
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Paul T Fanta
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Richard B Schwab
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Rebecca Shatsky
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Steven C Plaxe
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Andrew Sharabi
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
- Department of Radiation Medicine and Applied Sciences, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Edward Stites
- Integrative Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jacob J Adashek
- Department of Internal Medicine, University of South Florida, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Ryosuke Okamura
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Suzanna Lee
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Scott M Lippman
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Jason K Sicklick
- Center for Personalized Cancer Therapy and Division of Surgical Oncology, Department of Surgery, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| |
Collapse
|
26
|
Charo LM, Eskander RN, Okamura R, Patel SP, Nikanjam M, Lanman RB, Piccioni DE, Kato S, McHale MT, Kurzrock R. Clinical implications of plasma circulating tumor DNA in gynecologic cancer patients. Mol Oncol 2020; 15:67-79. [PMID: 32881280 PMCID: PMC7782073 DOI: 10.1002/1878-0261.12791] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/21/2020] [Accepted: 08/27/2020] [Indexed: 12/17/2022] Open
Abstract
Molecular characterization of cancers is important in dictating prognostic factors and directing therapy. Next‐generation sequencing of plasma circulating tumor DNA (ctDNA) offers less invasive, more convenient collection, and a more real‐time representation of a tumor and its molecular heterogeneity than tissue. However, little is known about the clinical implications of ctDNA assessment in gynecologic cancer. We describe the molecular landscape identified on ctDNA, ctDNA concordance with tissue‐based analysis, and factors associated with overall survival (OS) in gynecologic cancer patients with ctDNA analysis. We reviewed clinicopathologic and genomic information for 105 consecutive gynecologic cancer patients with ctDNA analysis, including 78 with tissue‐based sequencing, enrolled in the Profile‐Related Evidence Determining Individualized Cancer Therapy (NCT02478931) trial at the University of California San Diego Moores Cancer Center starting July 2014. Tumors included ovarian (47.6%), uterine (35.2%), cervical (12.4%), vulvovaginal (2.9%), and unknown gynecologic primary (1.9%). Most ovarian and uterine cancers (86%) were high grade. 34% (N = 17) of ovarian cancers had BRCA alterations, and 22% (N = 11) were platinum sensitive. Patients received median 2 (range 0–13) lines of therapy prior to ctDNA collection. Most (75.2%) had at least one characterized alteration on ctDNA analysis, and the majority had unique genomic profiles on ctDNA. Most common alterations were TP53 (N = 59, 56.2% of patients), PIK3CA (N = 26, 24.8%), KRAS (N = 14, 13.3%), BRAF (N = 10, 9.5%), ERBB2 (N = 8, 7.6%), and MYC (N = 8, 7.6%). Higher ctDNA maximum mutation allele frequency was associated with worse OS [hazard ratio (HR): 1.91, P = 0.03], while therapy matched to ctDNA alterations (N = 33 patients) was independently associated with improved OS (HR: 0.34, P = 0.007) compared to unmatched therapy (N = 28 patients) in multivariate analysis. Tissue and ctDNA genomic results showed high concordance unaffected by temporal or spatial factors. This study provides evidence for the utility of ctDNA in determining outcome and individualizing cancer therapy in patients with gynecologic cancer.
Collapse
Affiliation(s)
- Lindsey M Charo
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Ramez N Eskander
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Ryosuke Okamura
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Sandip P Patel
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Mina Nikanjam
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | | | - David E Piccioni
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Michael T McHale
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| |
Collapse
|
27
|
Graziano F, Fischer NW, Bagaloni I, Di Bartolomeo M, Lonardi S, Vincenzi B, Perrone G, Fornaro L, Ongaro E, Aprile G, Bisonni R, Prisciandaro M, Malkin D, Gariépy J, Fassan M, Loupakis F, Sarti D, Del Prete M, Catalano V, Alessandroni P, Magnani M, Ruzzo A. TP53 Mutation Analysis in Gastric Cancer and Clinical Outcomes of Patients with Metastatic Disease Treated with Ramucirumab/Paclitaxel or Standard Chemotherapy. Cancers (Basel) 2020; 12:2049. [PMID: 32722340 PMCID: PMC7465166 DOI: 10.3390/cancers12082049] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/14/2020] [Accepted: 07/22/2020] [Indexed: 12/16/2022] Open
Abstract
Loss of p53 promotes vascular endothelial growth factor (VEGF)-A up-regulation and the angiogenic potential of cancer cells. We investigated TP53 somatic mutations in 110 primary gastric adenocarcinomas of two retrospective metastatic series including 48 patients treated with second-line Ramucirumab/Paclitaxel and 62 patients who received first-line chemotherapy with Cisplatin or Oxaliplatin plus 5-Fluorouracil. Missense mutations were classified by tumor protein p53 (TP53) mutant-specific residual transcriptional activity scores (TP53RTAS) and used to stratify patients into two groups: transcriptionally TP53Active and TP53Inactive. The primary endpoint was overall survival (OS). An additional analysis was addressed to measure VEGF/VEGF receptor 2 (VEGFR2) expression levels in relation to the TP53RTAS. In the Ramucirumab/Paclitaxel group, 29/48 (60.4%) patients had TP53 mutations. Ten patients with TP53Inactive mutations showed better OS than carriers of other TP53 mutations. This effect was retained in the multivariate model analysis (Hazard Ratio = 0.29, 95% confidence interval = 0.17-0.85, p = 0.02). In the chemotherapy group, 41/62 (66%) patients had TP53 mutations, and the 11 carriers of TP53Inactive mutations showed the worst OS (Hazard Ratio = 2.64, 95% confidence interval = 1.17-5.95, p = 0.02). VEGF-A mRNA expression levels were significantly increased in TP53Inactive cases. Further studies are warranted to explore the effect of TP53Inactive mutations in different anti-cancer regimens. This information would lead to new tailored therapy strategies for this lethal disease.
Collapse
Affiliation(s)
- Francesco Graziano
- Medical Oncology Unit, Azienda, Ospedali Riuniti Marche Nord, 61121 Pesaro, Italy; (D.S.); (V.C.); (P.A.)
| | - Nicholas W. Fischer
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; (N.W.F.); (D.M.)
| | - Irene Bagaloni
- Department of Biomolecular Sciences, Università degli Studi di Urbino, 61032 Fano, Italy; (I.B.); (M.M.)
| | - Maria Di Bartolomeo
- Department of Medical Oncology, Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy; (M.D.B.); (M.P.)
| | - Sara Lonardi
- Veneto Institute of Oncology IOV–IRCCS, 35128 Padova, Italy; (S.L.); (M.F.); (F.L.)
| | - Bruno Vincenzi
- Department of Oncology, Campus Bio-Medico University, 00128 Rome, Italy; (B.V.); (G.P.)
| | - Giuseppe Perrone
- Department of Oncology, Campus Bio-Medico University, 00128 Rome, Italy; (B.V.); (G.P.)
| | - Lorenzo Fornaro
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, 56126 Pisa, Italy;
| | - Elena Ongaro
- Department of Oncology, University and General Hospital, 33100 Udine, Italy;
- Unit of Medical Oncology and Cancer Prevention, Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Giuseppe Aprile
- Department of Oncology, San Bortolo General Hospital, 36100 Vicenza, Italy;
| | - Renato Bisonni
- Medical Oncology Unit, Hospital of Fermo, 63900 Fermo, Italy; (R.B.); (M.D.P.)
| | - Michele Prisciandaro
- Department of Medical Oncology, Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy; (M.D.B.); (M.P.)
| | - David Malkin
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; (N.W.F.); (D.M.)
- Division of Hematology-Oncology, Department of Pediatrics, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada;
| | - Jean Gariépy
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada;
- Physical Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Matteo Fassan
- Veneto Institute of Oncology IOV–IRCCS, 35128 Padova, Italy; (S.L.); (M.F.); (F.L.)
| | - Fotios Loupakis
- Veneto Institute of Oncology IOV–IRCCS, 35128 Padova, Italy; (S.L.); (M.F.); (F.L.)
| | - Donatella Sarti
- Medical Oncology Unit, Azienda, Ospedali Riuniti Marche Nord, 61121 Pesaro, Italy; (D.S.); (V.C.); (P.A.)
| | - Michela Del Prete
- Medical Oncology Unit, Hospital of Fermo, 63900 Fermo, Italy; (R.B.); (M.D.P.)
| | - Vincenzo Catalano
- Medical Oncology Unit, Azienda, Ospedali Riuniti Marche Nord, 61121 Pesaro, Italy; (D.S.); (V.C.); (P.A.)
| | - Paolo Alessandroni
- Medical Oncology Unit, Azienda, Ospedali Riuniti Marche Nord, 61121 Pesaro, Italy; (D.S.); (V.C.); (P.A.)
| | - Mauro Magnani
- Department of Biomolecular Sciences, Università degli Studi di Urbino, 61032 Fano, Italy; (I.B.); (M.M.)
| | - Annamaria Ruzzo
- Department of Biomolecular Sciences, Università degli Studi di Urbino, 61032 Fano, Italy; (I.B.); (M.M.)
| |
Collapse
|
28
|
Fang S, Cheng W, Zhang M, Yang R. Association of TP53 Mutations with Response to Anlotinib Treatment in Advanced Non-Small Cell Lung Cancer. Onco Targets Ther 2020; 13:6645-6650. [PMID: 32753898 PMCID: PMC7354100 DOI: 10.2147/ott.s257052] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/15/2020] [Indexed: 12/24/2022] Open
Abstract
Multitargeted antiangiogenic drugs have demonstrated significant antitumor activity against a variety of solid tumors. Anlotinib, a novel oral multitargeted antiangiogenic tyrosine kinase inhibitor, was approved as a third-line treatment for advanced NSCLC in China. However, predictive biomarkers are currently insufficient and are urgently required. Herein, we report three pre-treated cases of advanced NSCLC with TP53 mutations, wherein these patients showed partial response to anlotinib. Moreover, the three patients have achieved a progression-free survival of 8, 6.5, and 5 months, respectively. The main toxicities were hypertension, hand-foot syndrome and fatigue. In conclusion, TP53 mutations may represent a biomarker for predicting salutary effects of anlotinib.
Collapse
Affiliation(s)
- Shencun Fang
- Department of Respiratory Medicine, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Wanwan Cheng
- Department of Respiratory Medicine, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Meiling Zhang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Rusong Yang
- Department of Thoracic Surgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
29
|
Kato S, Okamura R, Sicklick JK, Daniels GA, Hong DS, Goodman A, Weihe E, Lee S, Khalid N, Collier R, Mareboina M, Riviere P, Whitchurch TJ, Fanta PT, Lippman SM, Kurzrock R. Prognostic implications of RAS alterations in diverse malignancies and impact of targeted therapies. Int J Cancer 2020; 146:3450-3460. [PMID: 31782524 DOI: 10.1002/ijc.32813] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/26/2019] [Accepted: 10/09/2019] [Indexed: 02/03/2023]
Abstract
RAS alterations are often found in difficult-to-treat malignancies and are considered "undruggable." To better understand the clinical correlates and coaltered genes of RAS alterations, we used targeted next-generation sequencing (NGS) to analyze 1,937 patients with diverse cancers. Overall, 20.9% of cancers (405/1,937) harbored RAS alterations. Most RAS-altered cases had genomic coalterations (95.3%, median: 3, range: 0-51), often involving genes implicated in oncogenic signals: PI3K pathway (31.4% of 405 cases), cell cycle (31.1%), tyrosine kinase families (21.5%) and MAPK signaling (18.3%). Patients with RAS-altered versus wild-type RAS malignancies had significantly worse overall survival (OS; p = 0.02 [multivariate]), with KRAS alterations, in particular, showing shorter survival. Moreover, coalterations in both RAS and PI3K signaling or cell-cycle-associated genes correlated with worse OS (p = 0.004 and p < 0.0001, respectively [multivariate]). Among RAS-altered patients, MEK inhibitors alone did not impact progression-free survival (PFS), while matched targeted therapy against non-MAPK pathway coalterations alone showed a trend toward longer PFS (vs. patients who received unmatched therapy) (HR: 0.79, 95% CI: 0.61-1.03, p = 0.07). Three of nine patients (33%) given tailored combination therapies targeting both MAPK and non-MAPK pathways achieved objective responses. In conclusion, RAS alterations correlated with poor survival across cancers. The majority of RAS alterations were accompanied by coalterations impacting other oncogenic pathways. MEK inhibitors alone were ineffective against RAS-altered cancers while matched targeted therapy against coalterations alone correlated with a trend toward improved PFS. A subset of the small number of patients given MEK inhibitors plus tailored non-MAPK-targeting agents showed responses, suggesting that customized combinations warrant further investigation.
Collapse
Affiliation(s)
- Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, California
| | - Ryosuke Okamura
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, California
| | - Jason K Sicklick
- Center for Personalized Cancer Therapy and Division of Surgical Oncology, Department of Surgery, UC San Diego Moores Cancer Center, La Jolla, California
| | - Gregory A Daniels
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, California
| | - David S Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aaron Goodman
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, California
| | - Elizabeth Weihe
- Department of Radiology, UC San Diego Moores Cancer Center, La Jolla, California
| | - Suzanna Lee
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, California
| | - Noor Khalid
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, California
| | - Rachel Collier
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, California
| | - Manvita Mareboina
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, California
| | - Paul Riviere
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, California
| | - Theresa J Whitchurch
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, California
| | - Paul T Fanta
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, California
| | - Scott M Lippman
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, California
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, California
| |
Collapse
|
30
|
Caruso C, Garofalo C. Pharmacogenomics Biomarkers of Soft Tissue Sarcoma Therapies. Front Oncol 2020; 10:509. [PMID: 32351891 PMCID: PMC7174622 DOI: 10.3389/fonc.2020.00509] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 03/20/2020] [Indexed: 12/19/2022] Open
Abstract
Soft tissue sarcomas (STS) are heterogeneous rare malignancies comprising ~1% of all solid cancers in adults and including more than 70 histological and molecular subtypes with different pathological and clinical development characteristics. Over the last two decades, the increased knowledge of the new molecular and genomic mechanisms of different STS histotypes allowed for a reclassification of these tumors and consequently to the development of novel chemotherapeutic agents. Generally, surgery, in combination with radiotherapy only in selected cases of localized disease, represents the most common treatment of primary STS, whereas the principal treatment modality for locally advanced or metastatic disease is first-line chemotherapy. The principal treatment for the preponderance of STS patients is usually an anthracycline (epirubicin and doxorubicin) in monotherapy or in combination with other drug novel chemotherapeutic agents. However, survival for treated patients with metastatic disease is poor, and a 2-years survival rate is about 30%. In this scenario, Pharmacogenomics (PGx) biomarkers that can predict drug response play an important role in the improvement of molecular diagnostics in clinical routines and contribute to elucidating the genetic basis for the differences in treatment efficacy and toxicity among STS patients. This review focuses on recent insight in the PGx biomarkers that have been described to modulate responsiveness and toxicity parameters of conventional and new chemotherapeutics drugs in several STS histotypes.
Collapse
Affiliation(s)
| | - Cecilia Garofalo
- Advanced Translational Research Laboratory, Veneto Institute of Oncology IOV – IRCCS, Padua, Italy
| |
Collapse
|
31
|
Zhu Z, Jin Z, Zhang H, Zhang M, Sun D. Integrative Clustering Reveals a Novel Subtype of Soft Tissue Sarcoma With Poor Prognosis. Front Genet 2020; 11:69. [PMID: 32127798 PMCID: PMC7038822 DOI: 10.3389/fgene.2020.00069] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 01/21/2020] [Indexed: 12/13/2022] Open
Abstract
Background Soft tissue sarcomas (STSs) are heterogeneous at the clinical and molecular level and need to be further sub-clustered for treatment and prognosis. Materials And Methods STSs were sub-clustered based on RNAseq and miRNAseq data extracted from The Cancer Genome Atlas (TCGA) through the combined process of similarity network fusion (SNF) and consensus clustering (CC). The expression and clinical characteristics of each sub-cluster were analyzed. The genes differentially expressed (lncRNAs, miRNAs, and mRNAs) between the poor prognosis and good prognosis clusters were used to construct a competing endogenous RNA (ceRNA) network. Functional enrichment analysis was conducted and a hub network was extracted from the constructed ceRNA network. Results A total of 247 STSs were classified into three optimal sub-clusters, and patients in cluster 2 (C2) had a significantly lower rate of survival. A ceRNA network with 91 nodes and 167 edges was constructed according to the hypothesis of ceRNA. Functional enrichment analysis revealed that the network was mainly associated with organism development functions. Moreover, LncRNA (KCNQ1OT1)-miRNA (has-miR-29c-3p)-mRNA (JARID2, CDK8, DNMT3A, TET1)-competing endogenous gene pairs were identified as hub networks of the ceRNA network, in which each component showed survival significance. Conclusion Integrative clustering analysis revealed that the STSs could be clustered into three sub-clusters. The ceRNA network, especially the subnetwork LncRNA (KCNQ1OT1)-miRNA (has-miR-29c-3p)-mRNA (JARID2, CDK8, DNMT3A, TET1) was a promising therapeutic target for the STS sub-cluster associated with a poor prognosis.
Collapse
Affiliation(s)
- Zhenhua Zhu
- Department of Orthopaedic Trauma, The First Hospital of Jilin University, Changchun, China
| | - Zheng Jin
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Haibo Zhang
- College of Chemistry, Jilin University, Changchun, China
| | - Mei Zhang
- College of Chemistry, Jilin University, Changchun, China
| | - Dahui Sun
- Department of Orthopaedic Trauma, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
32
|
Kaseb AO, Sánchez NS, Sen S, Kelley RK, Tan B, Bocobo AG, Lim KH, Abdel-Wahab R, Uemura M, Pestana RC, Qiao W, Xiao L, Morris J, Amin HM, Hassan MM, Rashid A, Banks KC, Lanman RB, Talasaz A, Mills-Shaw KR, George B, Haque A, Raghav KPS, Wolff RA, Yao JC, Meric-Bernstam F, Ikeda S, Kurzrock R. Molecular Profiling of Hepatocellular Carcinoma Using Circulating Cell-Free DNA. Clin Cancer Res 2019; 25:6107-6118. [PMID: 31363003 PMCID: PMC9292132 DOI: 10.1158/1078-0432.ccr-18-3341] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 02/15/2019] [Accepted: 07/25/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Molecular profiling has been used to select patients for targeted therapy and determine prognosis. Noninvasive strategies are critical to hepatocellular carcinoma (HCC) given the challenge of obtaining liver tissue biopsies. EXPERIMENTAL DESIGN We analyzed blood samples from 206 patients with HCC using comprehensive genomic testing (Guardant Health) of circulating tumor DNA (ctDNA). RESULTS A total of 153/206 (74.3%) were men; median age, 62 years (range, 18-91 years). A total of 181/206 patients had ≥1 alteration. The total number of alterations was 680 (nonunique); median number of alterations/patient was three (range, 1-13); median mutant allele frequency (% cfDNA), 0.49% (range, 0.06%-55.03%). TP53 was the common altered gene [>120 alterations (non-unique)] followed by EGFR, MET, ARID1A, MYC, NF1, BRAF, and ERBB2 [20-38 alterations (nonunique)/gene]. Of the patients with alterations, 56.9% (103/181) had ≥1 actionable alterations, most commonly in MYC, EGFR, ERBB2, BRAF, CCNE1, MET, PIK3CA, ARID1A, CDK6, and KRAS. In these genes, amplifications occurred more frequently than mutations. Hepatitis B (HBV)-positive patients were more likely to have ERBB2 alterations, 35.7% (5/14) versus 8.8% HBV-negative (P = 0.04). CONCLUSIONS This study represents the first large-scale analysis of blood-derived ctDNA in HCC in United States. The genomic distinction based on HCC risk factors and the high percentage of potentially actionable genomic alterations suggests potential clinical utility for this technology.
Collapse
Affiliation(s)
- Ahmed O Kaseb
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Nora S Sánchez
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shiraj Sen
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robin K Kelley
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Benjamin Tan
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Andrea G Bocobo
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Kian H Lim
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Reham Abdel-Wahab
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Arizona Clinical Oncology Department, Assiut University Hospital, Assiut, Egypt
| | - Marc Uemura
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Wei Qiao
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lianchun Xiao
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffrey Morris
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hesham M Amin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Manal M Hassan
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Asif Rashid
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | | | - Kenna R Mills-Shaw
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bhawana George
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Abedul Haque
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kanwal P S Raghav
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert A Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - James C Yao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Funda Meric-Bernstam
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sadakatsu Ikeda
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego, Moores Cancer Center, La Jolla, California
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego, Moores Cancer Center, La Jolla, California.
| |
Collapse
|
33
|
Li AM, Boichard A, Kurzrock R. Mutated TP53 is a marker of increased VEGF expression: analysis of 7,525 pan-cancer tissues. Cancer Biol Ther 2019; 21:95-100. [PMID: 31564192 PMCID: PMC7012180 DOI: 10.1080/15384047.2019.1665956] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 05/20/2019] [Accepted: 09/01/2019] [Indexed: 01/05/2023] Open
Abstract
Anti-angiogenic therapies are an important class of anti-cancer treatment drugs. However, their efficacy is limited to certain tumors and would benefit from identifying a biomarker predictive of therapeutic response. TP53 (tumor protein p53) is a tumor suppressor gene frequently mutated in cancer and implicated in cell-cycle regulation, apoptosis, and angiogenesis. Data from 7,525 unique tumor samples (representing 30 tumor cohorts) were retrieved from the TCGA database to analyze the relationship between TP53-mutation status and VEGFA (vascular endothelial growth factor A) expression. Univariate analyses were done using a Mann-Whitney univariate test or Fisher's exact test. Parameters with a p-value (p)≤0.1 in univariate analysis were selected for follow-up multivariate analyses, including TP53-mutation status, cancer cohorts, cancer subtypes, and VEGFA expression. Our analysis demonstrates statistically significant increases in VEGFA mRNA tissue expression in TP53-mutated adenocarcinomas (but not in squamous cancers) compared to TP53 wild-type tumors. This association holds true in multivariate analyses and remains independent of HIF-1α and MDM2 overexpression. Our findings provide additional evidence that TP53 mutations are linked to the VEGF pathway, potentially offering insight into the mechanism behind increased sensitivity to anti-angiogenic therapies observed in some TP53-mutant tumors.
Collapse
Affiliation(s)
- Alex M. Li
- University of California San Diego School of Medicine, La Jolla, CA, USA
| | - Amélie Boichard
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| |
Collapse
|
34
|
Lee ATJ, Jones RL, Huang PH. Pazopanib in advanced soft tissue sarcomas. Signal Transduct Target Ther 2019; 4:16. [PMID: 31123606 PMCID: PMC6522548 DOI: 10.1038/s41392-019-0049-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 04/08/2019] [Accepted: 04/16/2019] [Indexed: 12/18/2022] Open
Abstract
Pazopanib is the first and only tyrosine kinase inhibitor currently approved for the treatment of multiple histological subtypes of soft tissue sarcoma (STS). Initially developed as a small molecule inhibitor of vascular endothelial growth factor receptors, preclinical work indicates that pazopanib exerts an anticancer effect through the inhibition of both angiogenic and oncogenic signaling pathways. Following the establishment of optimal dosing and safety profiles in early phase studies and approval for the treatment of advanced renal cell carcinoma, pazopanib was investigated in STS. A landmark phase III randomized study demonstrated improved progression-free survival with pazopanib compared to that with placebo in pretreated patients with STS of various subtypes. The efficacy of pazopanib in specific STS subtypes has been further described in real-world-based case series in both mixed and subtype-specific STS cohorts. At present, there are no clinically validated predictive biomarkers for use in selecting patients with advanced STS for pazopanib therapy, limiting the clinical effectiveness and cost-effectiveness of the drug. In this review, we summarize the preclinical and clinical data for pazopanib, outline the evidence base for its effect in STS and explore reported studies that have investigated putative biomarkers.
Collapse
Affiliation(s)
- Alex T. J. Lee
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - Robin L. Jones
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust, London, UK
- Division of Clinical Studies, Institute of Cancer Research, London, UK
| | - Paul H. Huang
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| |
Collapse
|
35
|
Carmagnani Pestana R, Groisberg R, Roszik J, Subbiah V. Precision Oncology in Sarcomas: Divide and Conquer. JCO Precis Oncol 2019; 3:PO.18.00247. [PMID: 32914012 PMCID: PMC7446356 DOI: 10.1200/po.18.00247] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2019] [Indexed: 12/18/2022] Open
Abstract
Sarcomas are a heterogeneous group of rare malignancies that exhibit remarkable heterogeneity, with more than 50 subtypes recognized. Advances in next-generation sequencing technology have resulted in the discovery of genetic events in these mesenchymal tumors, which in addition to enhancing understanding of the biology, have opened up avenues for molecularly targeted therapy and immunotherapy. This review focuses on how incorporation of next-generation sequencing has affected drug development in sarcomas and strategies for optimizing precision oncology for these rare cancers. In a significant percentage of soft tissue sarcomas, which represent up to 40% of all sarcomas, specific driver molecular abnormalities have been identified. The challenge to evaluate these mutations across rare cancer subtypes requires the careful characterization of these genetic alterations to further define compelling drivers with therapeutic implications. Novel models of clinical trial design also are needed. This shift would entail sustained efforts by the sarcoma community to move from one-size-fits-all trials, in which all sarcomas are treated similarly, to divide-and-conquer subtype-specific strategies.
Collapse
Affiliation(s)
| | - Roman Groisberg
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jason Roszik
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Vivek Subbiah
- The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
36
|
Mirili C, Paydaş S, Guney IB, Ogul A, Gokcay S, Buyuksimsek M, Yetisir AE, Karaalioglu B, Tohumcuoglu M, Seydaoglu G. Assessment of potential predictive value of peripheral blood inflammatory indexes in 26 cases with soft tissue sarcoma treated by pazopanib: a retrospective study. Cancer Manag Res 2019; 11:3445-3453. [PMID: 31114378 PMCID: PMC6485039 DOI: 10.2147/cmar.s191199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose The aim of this study was to evaluate the prognostic and predictive value of neutrophil-to-lymphocyte ratio (NLR), derived neutrophil-to-lymphocyte ratio (DNLR), lymphocyte-to-monocyte ratio (LMR), and platelet-to-lymphocyte ratio (PLR) in soft tissue sarcoma (STS) cases treated with pazopanib. Materials and methods The study population included 26 STS cases treated with pazopanib for at least 3 months. NLR, DNLR, LMR, and PLR were evaluated at baseline, and at third month of therapy and also compared with response to pazopanib. Median measurements were taken as cutoff for NLR (4.8), DNLR (3.1), LMR (3.6), and PLR (195). The associations between these cutoff values and survival times (progression-free survival [PFS] and overall survival [OS]) were assessed by Kaplan–Meier curves and Cox proportional models. Results Patients with low pretreatment NLR and DNLR had longer OS (P=0.022, P=0.018), but low PLR was found to be associated only with longer OS. Additionally, decrease in NLR and DNLR after 3 months of therapy as compared with pretreatment measurements was found to be associated with an advantage for OS (P=0.021, P=0.010, respectively) and PFS (P=0.005, P=0.001, respectively). Response to pazopanib; changes in NLR, DNLR, LMR, and PLR; and >3 metastatic sites were found to be independent risk factors in univariate analysis, but NLR was the only independent risk factor in multivariate analysis. Conclusion Low pretreatment and decrease in NLR and DNLR values, and regression/stable disease after 3 months of pazopanib are predictive factors for longer OS and PFS.
Collapse
Affiliation(s)
- Cem Mirili
- Department of Medical Oncology, Çukurova University Faculty of Medicine, Adana, Turkey,
| | - Semra Paydaş
- Department of Medical Oncology, Çukurova University Faculty of Medicine, Adana, Turkey,
| | - Isa B Guney
- Department of Nuclear Medicine, Çukurova University Faculty of Medicine, Adana, Turkey
| | - Ali Ogul
- Department of Medical Oncology, Çukurova University Faculty of Medicine, Adana, Turkey,
| | - Serkan Gokcay
- Department of Medical Oncology, Çukurova University Faculty of Medicine, Adana, Turkey,
| | - Mahmut Buyuksimsek
- Department of Medical Oncology, Çukurova University Faculty of Medicine, Adana, Turkey,
| | - Abdullah E Yetisir
- Department of Medical Oncology, Çukurova University Faculty of Medicine, Adana, Turkey,
| | - Bilgin Karaalioglu
- Department of Medical Oncology, Çukurova University Faculty of Medicine, Adana, Turkey,
| | - Mert Tohumcuoglu
- Department of Medical Oncology, Çukurova University Faculty of Medicine, Adana, Turkey,
| | - Gulsah Seydaoglu
- Department of Bioistatistics, Çukurova University Faculty of Medicine, Adana, Turkey
| |
Collapse
|
37
|
Molecular profiling of cancer patients enables personalized combination therapy: the I-PREDICT study. Nat Med 2019; 25:744-750. [PMID: 31011206 PMCID: PMC6553618 DOI: 10.1038/s41591-019-0407-5] [Citation(s) in RCA: 450] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 02/22/2019] [Indexed: 12/17/2022]
Abstract
Cancer treatments have evolved from indiscriminate cytotoxic agents to
selective genome- and immune-targeted drugs that have transformed outcomes for
some malignancies.1 Tumor
complexity and heterogeneity suggest that the “precision medicine”
paradigm of cancer therapy requires treatment to be personalized to the
individual patient.2–6 To date, precision oncology
trials have been based upon molecular matching with predetermined
monotherapies.7–14 Several of these trials have
been hindered by very low matching rates, often in the 5–10%
range,15 and low
response rates. Low matching rates may be due to the use of limited gene panels,
restrictive molecular matching algorithms, lack of drug availability or the
deterioration and death of end-stage patients before therapy can be implemented.
We hypothesized that personalized treatment with combination therapies would
improve outcomes in patients with refractory malignancies. As a first test of
this concept, we implemented a cross-institutional, prospective study
(I-PREDICT, NCT02534675) that used tumor DNA sequencing and timely
recommendations for individualized treatment with combination therapies. We
found that administration of customized multi-drug regimens was feasible, with
49% of consented patients receiving personalized treatment. Targeting of a
larger fraction of identified molecular alterations, yielding a higher
“matching score,” was correlated with significantly improved
disease control rates, as well as longer progression-free and overall survival
rates, as compared to when fewer somatic alterations were targeted. Our findings
suggest that the current clinical trial paradigm for precision oncology, which
pairs one driver mutation with one drug, may be optimized by treating
molecularly complex and heterogeneous cancers with combinations of customized
agents.
Collapse
|
38
|
Shatsky R, Parker BA, Bui NQ, Helsten T, Schwab RB, Boles SG, Kurzrock R. Next-Generation Sequencing of Tissue and Circulating Tumor DNA: The UC San Diego Moores Center for Personalized Cancer Therapy Experience with Breast Malignancies. Mol Cancer Ther 2019; 18:1001-1011. [DOI: 10.1158/1535-7163.mct-17-1038] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 04/12/2018] [Accepted: 03/12/2019] [Indexed: 11/16/2022]
|
39
|
Nakano K, Funauchi Y, Hayakawa K, Tanizawa T, Ae K, Matsumoto S, Takahashi S. Relative Dose Intensity of Induction-Phase Pazopanib Treatment of Soft Tissue Sarcoma: Its Relationship with Prognoses of Pazopanib Responders. J Clin Med 2019; 8:jcm8010060. [PMID: 30626115 PMCID: PMC6352274 DOI: 10.3390/jcm8010060] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/20/2018] [Accepted: 01/04/2019] [Indexed: 01/07/2023] Open
Abstract
The approved standard dose of pazopanib is 800 mg per day, but the appropriate dose of pazopanib to treat soft tissue sarcoma (STS) patients in real-world practice is controversial. Of 124 STS patients treated with pazopanib, we retrospectively analyzed the cases of STS patients who achieved progression-free survival at 12 weeks by pazopanib treatment as pazopanib responders, and we evaluated their relative dose intensity (RDI) in the initial 12 weeks (12W-RDI). We enrolled 78 STS patients in the analyses as pazopanib responders, and 54 patients of the 78 pazopanib responders (69%) were able to maintain 12W-RDI ≥80%. In landmark analyses, patients with 12W-RDI of 80% ≥80% had significantly longer progression-free survival compared to those with 12W-RDI <80% (30.7 weeks vs. 22.0 weeks, hazard ratio [HR]: 0.56 [95%CI: 0.33–0.94], p = 0.026). The most frequently observed reasons of treatment interruption and/or dose reduction of pazopanib during the initial 12 weeks were anorexia and liver function disorders. Liver toxicity was the adverse event most frequently observed in the 12W-RDI <80% patients throughout the treatment periods. Based on our results, it appears that maintaining as high a dose intensity as possible that is tolerable—at least during the initial 12 weeks—is likely to be the better option in pazopanib treatment for STS patients.
Collapse
Affiliation(s)
- Kenji Nakano
- Department of Medical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan.
| | - Yuki Funauchi
- Department of Orthopedic Surgical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan.
| | - Keiko Hayakawa
- Department of Orthopedic Surgical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan.
| | - Taisuke Tanizawa
- Department of Orthopedic Surgical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan.
| | - Keisuke Ae
- Department of Orthopedic Surgical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan.
| | - Seiichi Matsumoto
- Department of Orthopedic Surgical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan.
| | - Shunji Takahashi
- Department of Medical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan.
| |
Collapse
|
40
|
Su YH, Kim AK, Jain S. Liquid biopsies for hepatocellular carcinoma. Transl Res 2018; 201:84-97. [PMID: 30056068 PMCID: PMC6483086 DOI: 10.1016/j.trsl.2018.07.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 06/18/2018] [Accepted: 07/02/2018] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is the world's second leading cause of cancer death; 82.4% of patients die within 5 years. This grim prognosis is the consequence of a lack of effective early detection tools, limited treatment options, and the high frequency of HCC recurrence. Advances in the field of liquid biopsy hold great promise in improving early detection of HCC, advancing patient prognosis, and ultimately increasing the survival rate. In an effort to address the current challenges of HCC screening and management, several studies have identified and evaluated liver-cancer-associated molecular signatures such as genetic alterations, methylation, and noncoding RNA expression in the form of circulating biomarkers in body fluids and circulating tumor cells of HCC patients. In this review, we summarize the recent progress in HCC liquid biopsy, organized by the intended clinical application of the reported study.
Collapse
Affiliation(s)
- Ying-Hsiu Su
- The Baruch S. Blumberg Institute, Doylestown, Pennsylvania.
| | - Amy K Kim
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins School of Medicine, Baltimore Maryland.
| | - Surbhi Jain
- JBS Science, Inc., Doylestown, Pennsylvania.
| |
Collapse
|
41
|
Apatinib-based targeted therapy against pulmonary sarcomatoid carcinoma: a case report and literature review. Oncotarget 2018; 9:33734-33738. [PMID: 30263099 PMCID: PMC6154744 DOI: 10.18632/oncotarget.25989] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 03/17/2018] [Indexed: 12/15/2022] Open
Abstract
Sarcomatoid carcinoma is a rare malignancy characterized by a combination of epithelial and sarcoma or sarcoma-like components. In this study, we reported one case of pulmonary sarcomatoid carcinoma and evaluated the safety and efficacy of apatinib, a tyrosine kinase inhibitor selectively targeting vascular endothelial growth factor receptor 2, in treating this disease. The tumor mass was detected in the left lung of a 75-year-old man and showed positive immunostaining for cytokeratin (CK) 7, CK8, smooth muscle actin, CD31, and CD34. Next-generation sequencing analysis identified 4 mutations in NF1 (p.Q347Sfs*29), CDKN2A (p.G23V), ERBB3 (p.V104L), and TP53 (p.V157F) genes. The patient was given apatinib (250 mg) orally once per day. Sustained tumor regression was observed after apatinib treatment. There was no sever complication associated with apatinib therapy. In conclusion, apatinib-based targeted therapy may represent an important option for patients with sarcomatoid carcinoma.
Collapse
|
42
|
Kato S, Ross JS, Gay L, Dayyani F, Roszik J, Subbiah V, Kurzrock R. Analysis of MDM2 Amplification: Next-Generation Sequencing of Patients With Diverse Malignancies. JCO Precis Oncol 2018; 2018. [PMID: 30148248 PMCID: PMC6106866 DOI: 10.1200/po.17.00235] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purpose MDM2 amplification can promote tumorigenesis directly or indirectly through p53 inhibition. MDM2 has increasing clinical relevance because inhibitors are under evaluation in clinical trials, and MDM2 amplification is a possible genomic correlate of accelerated progression, known as hyperprogression, after anti–PD-1/PD-L1 immunotherapy. We used next-generation sequencing (NGS) to ascertain MDM2 amplification status across a large number of diverse cancers. Methods We interrogated the molecular profiles of 102,878 patients with diverse malignancies for MDM2 amplification and co-altered genes using clinical-grade NGS (182 to 465 genes). Results MDM2 amplification occurred in 3.5% of patients (3,650 of 102,878). The majority of tumor types had a small subset of patients with MDM2 amplification. Most of these patients (99.0% [3,613/3,650]) had co-alterations that accompanied MDM2 amplification. Various pathways, including those related to tyrosine kinase (37.9% [1,385 of 3,650]), PI3K signaling (25.4% [926 of 3,650]), TP53 (24.9% [910 of 3,650]), and MAPK signaling (23.6% [863 of 3,650]), were involved. Although infrequent, mismatch repair genes and PD-L1 amplification also were co-altered (2.2% [79 of 3,650]). Most patients (97.6% [3,563 of 3,650]) had one or more co-alterations potentially targetable with either a Food and Drug Administration–approved or investigational agent. MDM2 amplifications were less frequently associated with high tumor mutation burden compared with the MDM2 wild-type population (2.9% v 6.5%; P < .001). An illustrative patient who harbored MDM2 amplification and experienced hyperprogression with an immune checkpoint inhibitor is presented. Conclusion MDM2 amplification was found in 3.5% of 102,878 patients, 97.6% of whom harbored genomic co-alterations that were potentially targetable. This study suggests that a small subset of most tumor types have MDM2 amplification as well as pharmacologically tractable co-alterations.
Collapse
Affiliation(s)
- Shumei Kato
- University of California, San Diego, Moores Cancer Center, La Jolla
| | | | | | | | - Jason Roszik
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Vivek Subbiah
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Razelle Kurzrock
- University of California, San Diego, Moores Cancer Center, La Jolla
| |
Collapse
|
43
|
Lu H, Yang S, Zhu H, Tong X, Xie F, Qin J, Han N, Wu X, Fan Y, Shao YW, Mao W. Targeted next generation sequencing identified clinically actionable mutations in patients with esophageal sarcomatoid carcinoma. BMC Cancer 2018; 18:251. [PMID: 29506494 PMCID: PMC5838991 DOI: 10.1186/s12885-018-4159-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 02/21/2018] [Indexed: 02/08/2023] Open
Abstract
Background Esophageal sarcomatoid carcinoma (ESC) is a rare disease with a mixture of both carcinomatous and sarcomatous components in the tumor. Its genetic background and mechanisms of oncogenesis remain largely unknown. Methods Here we performed targeted next generation sequencing (NGS) on a pan-cancer gene panel in 15 ESC tumors to explore their genetic alterations, and aimed to identify clinically actionable mutations for future treatment instructions. Results TP53 alterations were identified in all patients. Alterations in receptor tyrosine kinases (RTK) were identified in 10 out of 15 patients. Members of downstream RAS and PI3-kinase pathways are also mutated in 10 patients, and PIK3CA is the top mutated gene in these pathways. In addition, we identified mutations on histone modification genes in 5 patients, including histone acetyltransferase gene EP300 and its homologue CREBBP, lysine methyltransferase genes KMT2A and KMT2B, and lysine demethylase gene KDM5A. Finally, mismatch repair (MMR) genes and proofreading gene POLE all together were mutated in one third of the ESC patients. Conclusions This is the first study to unravel the mutational profile of ESC tumors. Our findings could match 9 patients to the targeted therapies currently available in clinical practice or in active clinical trials, suggesting the potential utility of targeted therapies for this rare disease in the future. Electronic supplementary material The online version of this article (10.1186/s12885-018-4159-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hongyang Lu
- Zhejiang Key Laboratory of Diagnosis & Treatment Technology on Thoracic Oncology (lung and esophagus), Zhejiang Cancer Hospital, NO.1 East Banshan Road, Gongshu District, Hangzhou, 310022, People's Republic of China.,Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, People's Republic of China
| | - Shifeng Yang
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, 310022, People's Republic of China
| | - Huineng Zhu
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, 310022, People's Republic of China
| | - Xiaoling Tong
- Translational Medicine Research Institute, Geneseeq Technology Inc, Suite 300, MaRS Centre, South Tower, 101 College Street, Toronto, ON, M5G 1L7, Canada
| | - Fajun Xie
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, People's Republic of China
| | - Jing Qin
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, People's Republic of China
| | - Na Han
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, People's Republic of China
| | - Xue Wu
- Translational Medicine Research Institute, Geneseeq Technology Inc, Suite 300, MaRS Centre, South Tower, 101 College Street, Toronto, ON, M5G 1L7, Canada
| | - Yun Fan
- Zhejiang Key Laboratory of Diagnosis & Treatment Technology on Thoracic Oncology (lung and esophagus), Zhejiang Cancer Hospital, NO.1 East Banshan Road, Gongshu District, Hangzhou, 310022, People's Republic of China.,Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, 310022, People's Republic of China
| | - Yang W Shao
- Translational Medicine Research Institute, Geneseeq Technology Inc, Suite 300, MaRS Centre, South Tower, 101 College Street, Toronto, ON, M5G 1L7, Canada.
| | - Weimin Mao
- Zhejiang Key Laboratory of Diagnosis & Treatment Technology on Thoracic Oncology (lung and esophagus), Zhejiang Cancer Hospital, NO.1 East Banshan Road, Gongshu District, Hangzhou, 310022, People's Republic of China.
| |
Collapse
|
44
|
Nakano K, Takahashi S. Current Molecular Targeted Therapies for Bone and Soft Tissue Sarcomas. Int J Mol Sci 2018; 19:E739. [PMID: 29510588 PMCID: PMC5877600 DOI: 10.3390/ijms19030739] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 03/01/2018] [Accepted: 03/03/2018] [Indexed: 12/16/2022] Open
Abstract
Systemic treatment options for bone and soft tissue sarcomas remained unchanged until the 2000s. These cancers presented challenges in new drug development partly because of their rarity and heterogeneity. Many new molecular targeting drugs have been tried in the 2010s, and some were approved for bone and soft tissue sarcoma. As one of the first molecular targeted drugs approved for solid malignant tumors, imatinib's approval as a treatment for gastrointestinal stromal tumors (GISTs) has been a great achievement. Following imatinib, other tyrosine kinase inhibitors (TKIs) have been approved for GISTs such as sunitinib and regorafenib, and pazopanib was approved for non-GIST soft tissue sarcomas. Olaratumab, the monoclonal antibody that targets platelet-derived growth factor receptor (PDGFR)-α, was shown to extend the overall survival of soft tissue sarcoma patients and was approved in 2016 in the U.S. as a breakthrough therapy. For bone tumors, new drugs are limited to denosumab, a receptor activator of nuclear factor κB ligand (RANKL) inhibitor, for treating giant cell tumors of bone. In this review, we explain and summarize the current molecular targeting therapies approved and in development for bone and soft tissue sarcomas.
Collapse
Affiliation(s)
- Kenji Nakano
- Department of Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo 135-8550, Japan.
| | - Shunji Takahashi
- Department of Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto, Tokyo 135-8550, Japan.
| |
Collapse
|
45
|
Ikeda S, Lim JS, Kurzrock R. Analysis of Tissue and Circulating Tumor DNA by Next-Generation Sequencing of Hepatocellular Carcinoma: Implications for Targeted Therapeutics. Mol Cancer Ther 2018; 17:1114-1122. [PMID: 29483209 DOI: 10.1158/1535-7163.mct-17-0604] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 11/21/2017] [Accepted: 02/12/2018] [Indexed: 12/15/2022]
Abstract
Hepatocellular carcinoma (HCC) has limited treatment options. Molecular analysis of its mutational landscape may enable the identification of novel therapies. However, biopsy is not routinely performed in HCC. The utility of analyzing cell-free circulating tumor DNA (ctDNA) by next-generation sequencing (NGS) is not established. We performed 32 ctDNA NGS analyses on 26 patients; 10 of these patients had tissue NGS (236 to 626 genes). ctDNA was evaluated using an assay that detects single nucleotide variants, amplifications, fusions, and specific insertion/deletion alterations in 54 to 70 genes. The ctDNA demonstrated that 23 of 26 patients (88.5%) had ≥1 characterized alteration, and all these individuals had ≥1 potentially actionable alteration. The most frequently mutated gene was TP53 (16 of 26 patients, 61.5%). There were 47 unique characterized molecular alterations among 18 total gene alterations [variants of unknown significance (VUS) excluded)]. ctDNA and tissue NGS frequently showed different profiles, perhaps due to length of time between tissue and blood samples [median = 370 days (range, 29 to 876 days)]. Serial ctDNA evaluation in an illustrative patient treated with capecitabine demonstrated emergence of a new TP53 alteration after progression. In conclusion, ctDNA profiling is feasible in advanced HCC, and serial assessment using ctDNA NGS can reveal genomic changes with time. NGS of ctDNA provides a minimally invasive alternative for identifying potentially actionable gene alterations and potential molecular targeted therapies. Dynamic changes in molecular portfolio associated with therapeutic pressure in difficult-to-biopsy patients can be observed. Mol Cancer Ther; 17(5); 1114-22. ©2018 AACR.
Collapse
Affiliation(s)
- Sadakatsu Ikeda
- Center for Personalized Cancer Therapy, Division of Hematology/Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla, California. .,Tokyo Medical and Dental University, Tokyo, Japan
| | - Jordan S Lim
- Center for Personalized Cancer Therapy, Division of Hematology/Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla, California.
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy, Division of Hematology/Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla, California
| |
Collapse
|
46
|
Monga V, Swami U, Tanas M, Bossler A, Mott SL, Smith BJ, Milhem M. A Phase I/II Study Targeting Angiogenesis Using Bevacizumab Combined with Chemotherapy and a Histone Deacetylase Inhibitor (Valproic Acid) in Advanced Sarcomas. Cancers (Basel) 2018; 10:E53. [PMID: 29462961 PMCID: PMC5836085 DOI: 10.3390/cancers10020053] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 12/18/2022] Open
Abstract
Epigenetic events and genetic alterations under the control of the tumor microenvironment potentially mediate tumor induced angiogenesis involved in soft tissue sarcoma (STS) metastasis. Addition of antiangiogenic agent, such as bevacizumab, to standard chemotherapy in treatment of sarcoma has been studied in clinical trials, but most of the findings have not supported its use. We hypothesized the existence of an epigenetically mediated "angiogenic switch", and the tumor microenvironment, prevents bevacizumab from truly blocking angiogenesis. The addition of valproic acid (VPA), a weak histone deacetylase inhibitor, and bevacizumab, a monoclonal antibody against vascular endothelial growth factor, together with the cytotoxic effects of gemcitabine and docetaxel, may enhance responses and alter chemoresistance. This was designed as a phase I/II trial with primary endpoints including safety of the treatment combination and tumor response. Unresectable or metastatic sarcoma patients >18 years of age, irrespective of number of prior treatments, received VPA 40 mg/kg orally for 5 days prior to day 1, bevacizumab at 15 mg/kg IV on day 1, gemcitabine 900 mg/m² (day 1, day 8), and docetaxel 75 mg/m² (day 8). Cycles were of 28 day duration. Bevacizumab and VPA were continued as maintenance after 6 cycles, until disease progression. A standard 3 + 3 phase I dose de-escalation design was utilized to evaluate safety. Gain of function p53 gene mutation testing was performed on available archival tissue specimens. A total of 46 patients (30 female, 16 male) with median age of 60 (range 24-81) years were enrolled; 34 (73.9%) patients received prior chemotherapy, 14 (30%) of which received prior gemcitabine and docetaxel. Patients received a median of 5.5 cycles (range 0-24 of treatment (min 0, one patient died prior to completing the first cycle; max: 24, one patient received 6 cycles and 18 maintenance cycles before progressing). Seventeen patients underwent dose reduction, of which VPA was reduced in 6 patients. Forty-one patients were evaluable for response. There was a confirmed complete response in 1 (epithelioid sarcoma), and a partial response (PR) in 6 (1 carcinosarcoma, 2 extrauterine leiomyosarcoma (LMS), 2 undifferentiated pleomorphic sarcoma, and 1 uterine LMS) patients. Stable disease (SD) was seen in 21 patients for at least 2 months. One subject with prior gemcitabine and docetaxel had PR, and 7 had SD. Median progression-free survival (PFS) was 5.7 months (95% CI: 2.1-8.0), and overall survival (OS) was 12.9 months (95% CI: 8.3-14.5). Three patients died due to tumor progression while on the study. The combination of VPA, bevacizumab, gemcitabine, and docetaxel appears to be moderately safe and well tolerated. Given that there are very limited options for patients with relapsed refractory STS, this drug combination may be an important therapy to consider. This combination treatment deserves further investigation in epithelioid and carcinosarcoma subtypes.
Collapse
Affiliation(s)
- Varun Monga
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| | - Umang Swami
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| | - Munir Tanas
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| | - Aaron Bossler
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| | - Sarah L Mott
- Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA.
| | - Brian J Smith
- Department of Biostatistics, University of Iowa College of Public Health & Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA.
| | - Mohammed Milhem
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| |
Collapse
|
47
|
Frequency of Somatic TP53 Mutations in Combination with Known Pathogenic Mutations in Colon Adenocarcinoma, Non-Small Cell Lung Carcinoma, and Gliomas as Identified by Next-Generation Sequencing. Neoplasia 2018; 20:256-262. [PMID: 29454261 PMCID: PMC5849803 DOI: 10.1016/j.neo.2017.12.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 12/14/2017] [Accepted: 12/18/2017] [Indexed: 12/20/2022] Open
Abstract
The tumor suppressor gene TP53 is the most frequently mutated gene in human cancer. It encodes p53, a DNA-binding transcription factor that regulates multiple genes involved in DNA repair, metabolism, cell cycle arrest, apoptosis, and senescence. TP53 is associated with human cancer by mutations that lead to a loss of wild-type p53 function as well as mutations that confer alternate oncogenic functions that enable them to promote invasion, metastasis, proliferation, and cell survival. Identifying the discrete TP53 mutations in tumor cells may help direct therapies that are more effective. In this study, we identified the frequency of individual TP53 mutations in patients with colon adenocarcinoma (48%), non–small cell lung carcinoma (NSCLC) (36%), and glioma/glioblastoma (28%) at our institution using next-generation sequencing. We also identified the occurrence of somatic mutations in numerous actionable genes including BRAF, EGFR, KRAS, IDH1, and PIK3CA that occurred concurrently with these TP53 mutations. Of the 480 tumors examined that contained one or more mutations in the TP53 gene, 219 were colon adenocarcinomas, 215 were NSCLCs, and 46 were gliomas/glioblastomas. Among the patients positive for TP53 mutations diagnosed with colon adenocarcinoma, 50% also showed at least one mutation in pathogenic genes of which 14% were BRAF, 33% were KRAS, and 3% were NRAS. Forty-seven percent of NSCLC patients harboring TP53 mutations also had a mutation in at least one actionable pathogenic variant with the following frequencies: BRAF: 4%, EGFR: 10%, KRAS: 28%, and PIK3CA: 4%. Fifty-two percent of patients diagnosed with glioma/glioblastoma with a positive TP53 mutation had at least one concurrent mutation in a known pathogenic gene of which 9% were CDKN2A, 41% were IDH1, and 11% were PIK3CA.
Collapse
|
48
|
Stacchiotti S, Van Tine BA. Synovial Sarcoma: Current Concepts and Future Perspectives. J Clin Oncol 2018; 36:180-187. [DOI: 10.1200/jco.2017.75.1941] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Synovial sarcoma (SS) is a rare sarcoma driven by a translocation between SS18 and SSX 1, 2, or 4. With approximately 800 to 1,000 cases a year in the United States, it most commonly affects young adults between the ages of 15 and 30 years. The resultant tumors are either monophasic (pure sarcomas), biphasic (a combination or epithelioid and sarcomatous components), or poorly differentiated. The hybrid transcription factor SS18:SSX alters SWItch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodeling and global methylation patterns that may allow for future therapeutic opportunities. In this review, we focus on the pharmacologic management of SS, both in the curative setting, where the standard approach is wide surgical excision combined with radiotherapy and/or (neo)adjuvant chemotherapy as appropriate, and in the palliative setting. In advanced disease, chemotherapy with anthracyclines and/or ifosfamide, trabectedin, or pazopanib has been demonstrated to be more active compared with other soft tissue sarcomas. In addition, a better understanding of the molecular and immunologic characteristics of SS has allowed for the identification of new potential targets and the development of novel biology-driven therapies that are all at different stages of testing. There include targeted agents, immunotherapy, and metabolic therapies. Because the impact of these strategies for improving SS outcome is still limited, current and future research is strongly needed to better understand the tumor biology, to identify predictive biomarkers, and to improve the outcomes for patients with SS.
Collapse
Affiliation(s)
- Silvia Stacchiotti
- Silvia Stacchiotti, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale Tumori, Milan, Italy; and Brian Andrew Van Tine, Washington University in St Louis, St Louis, MO
| | - Brian Andrew Van Tine
- Silvia Stacchiotti, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale Tumori, Milan, Italy; and Brian Andrew Van Tine, Washington University in St Louis, St Louis, MO
| |
Collapse
|
49
|
Goodman AM, Kato S, Cohen PR, Boichard A, Frampton G, Miller V, Stephens PJ, Daniels GA, Kurzrock R. Genomic landscape of advanced basal cell carcinoma: Implications for precision treatment with targeted and immune therapies. Oncoimmunology 2017; 7:e1404217. [PMID: 29399405 PMCID: PMC5790366 DOI: 10.1080/2162402x.2017.1404217] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/06/2017] [Accepted: 11/08/2017] [Indexed: 01/16/2023] Open
Abstract
Metastatic basal cell cancer (BCC) is an ultra-rare malignancy with no approved therapies beyond Hedgehog inhibitors. We characterized the genomics, tumor mutational burden (TMB), and anti-PD-1 therapy responses in patients with locally advanced or metastatic BCC. Overall, 2,039 diverse cancer samples that had undergone comprehensive genomic profiling (CGP) were reviewed. Eight patients with locally advanced/metastatic BCC were identified (two had two CGP analyses; total, 10 biopsies). Two tumors demonstrated PD-L1 amplification. Seven patients had >1 actionable alteration. The TMB (mutations/mb) (median (range)) was 90 (3-103) for the BCCs versus 4 (1-860) for 1637 cancers other than BCC (P < 0.0001). Median progression-free survival (PFS) for all four patients treated with PD-1 blockade was 10.7 months (range, 3.8 to 17.6+ months); three patients had an objective response. In conclusion, advanced/metastatic BCC often has biological features (high TMB; PD-L1 amplification) predictive of immunotherapy benefit, and patients frequently respond to PD-1 blockade.
Collapse
Affiliation(s)
- Aaron M. Goodman
- Department of Medicine, Division of Hematology/Oncology, University of California San Diego, La Jolla, CA, USA
- Center for Personalized Cancer Therapy, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, Division of Blood and Marrow Transplantation, University of California San Diego, La Jolla, CA, USA
| | - Shumei Kato
- Department of Medicine, Division of Hematology/Oncology, University of California San Diego, La Jolla, CA, USA
- Center for Personalized Cancer Therapy, University of California San Diego, La Jolla, CA, USA
| | - Philip R. Cohen
- Department of Dermatology, University of California San Diego, La Jolla, CA, USA
| | - Amélie Boichard
- Center for Personalized Cancer Therapy, University of California San Diego, La Jolla, CA, USA
| | | | | | | | - Gregory A. Daniels
- Department of Medicine, Division of Hematology/Oncology, University of California San Diego, La Jolla, CA, USA
| | - Razelle Kurzrock
- Department of Medicine, Division of Hematology/Oncology, University of California San Diego, La Jolla, CA, USA
- Center for Personalized Cancer Therapy, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
50
|
Kobayashi H, Okuma T, Oka H, Hirai T, Ohki T, Ikegami M, Sawada R, Shinoda Y, Akiyama T, Sato K, Abe S, Kawano H, Goto T, Tanaka S. Neutrophil-to-lymphocyte ratio after pazopanib treatment predicts response in patients with advanced soft-tissue sarcoma. Int J Clin Oncol 2017; 23:368-374. [PMID: 29086877 DOI: 10.1007/s10147-017-1199-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 10/04/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND Pazopanib is a multi-tyrosine kinase inhibitor that is used to treat advanced soft-tissue sarcoma, and its efficacy has been confirmed in several clinical trials, although no clinically useful biomarkers have been identified. In other cancers, the neutrophil-to-lymphocyte ratio (NLR), the platelet-to-lymphocyte ratio (PLR), and the lymphocyte-to-monocyte ratio (LMR) are associated with chemotherapy response and prognosis. Therefore, we aimed to evaluate the associations of pazopanib response with NLR, PLR, and LMR among patients with advanced soft-tissue sarcoma. METHODS Data regarding NLR, PLR, and LMR were obtained for 25 patients who received pazopanib for soft-tissue sarcoma. The patients were categorized according to their values for NLR (≥3.8 vs. <3.8), PLR (≥230 vs. <230), and LMR (≥2.4 vs. <2.4), and we evaluated the associations of these markers with progression-free survival and overall survival using Kaplan-Meier curves and Cox proportional models. RESULTS No significant differences in progression-free survival or overall survival were observed based on the pre-treatment NLR, PLR, and LMR values. However, decreased NLR values after treatment using pazopanib were independently associated with significantly prolonged progression-free survival (hazard ratio: 0.07, p = 0.001) and overall survival (hazard ratio: 0.17, p = 0.0006). CONCLUSIONS Decreased NLR values after treatment using pazopanib may predict high efficacy and favorable outcomes among patients with advanced soft-tissue sarcoma.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Tomotake Okuma
- Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
| | - Hiroyuki Oka
- Department of Medical Research and Management for Musculoskeletal Pain, 22nd Century Medical and Research Center, Faculty of Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Toshihide Hirai
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Takahiro Ohki
- Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
| | - Masachika Ikegami
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Ryoko Sawada
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yusuke Shinoda
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Toru Akiyama
- Department of Orthopaedic Surgery, Saitama Medical Center, Jichi Medical University, 1-847 Amanuma, Omiya-ku, Saitama-shi, Saitama, 330-8503, Japan
| | - Kenji Sato
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Teikyo, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8606, Japan
| | - Satoshi Abe
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Teikyo, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8606, Japan
| | - Hirotaka Kawano
- Department of Orthopaedic Surgery, Faculty of Medicine, University of Teikyo, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8606, Japan
| | - Takahiro Goto
- Department of Musculoskeletal Oncology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-ku, Tokyo, 113-8677, Japan
| | - Sakae Tanaka
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|