1
|
Somasundaram E, Anderson PM, Smile TD, Halima A, Broughman JB, Reddy CA, Parsai S, Scott JG, Chan T, Campbell S, Angelov L, Zahler S, Trucco M, Thomas SM, Johnson S, Qi P, Magnelli A, Murphy ES. Neutrophil to lymphocyte ratio (NTLR) predicts local control and overall survival after stereotactic body radiotherapy (SBRT) in metastatic sarcoma. Sci Rep 2023; 13:19256. [PMID: 37935813 PMCID: PMC10630331 DOI: 10.1038/s41598-023-46476-3] [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: 02/10/2023] [Accepted: 11/01/2023] [Indexed: 11/09/2023] Open
Abstract
The neutrophil to lymphocyte ratio (NTLR) and absolute lymphocyte count (ALC) recovery are prognostic across many cancers. We investigated whether NLTR predicts SBRT success or survival in a metastatic sarcoma cohort treated with SBRT from 2014 and 2020 (N = 42). Wilcox Signed Rank Test and Friedman Test compare NTLR changes with local failure vs. local control (N = 138 lesions). Cox analyses identified factors associated with overall survival. If local control was successful, NLTR change was not significant (p = 0.30). However, NLTR significantly changed in patients with local failure (p = 0.027). The multivariable Cox model demonstrated higher NLTR before SBRT was associated with worse overall survival (p = 0.002). The optimal NTLR cut point was 5 (Youden index: 0.418). One-year overall survival in SBRT metastatic sarcoma cohort was 47.6% (CI 34.3%-66.1%). Patients with an NTLR above 5 had a one-year overall survival of 37.7% (21.4%-66.3%); patients with an NTLR below 5 had a significantly improved overall survival of 63% (43.3%-91.6%, p = 0.014). Since NTLR at the time of SBRT was significantly associated with local control success and overall survival in metastatic sarcoma treated with SBRT, future efforts to reduce tumor inhibitory microenvironment factors and improve lymphocyte recovery should be investigated.
Collapse
Affiliation(s)
| | - Peter M Anderson
- Department of Pediatric Hematology Oncology and Blood and Marrow Transplantation, Cleveland Clinic, Cleveland, OH, USA
| | - Timothy D Smile
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, R3 9500 Euclid Ave, Cleveland, 44195, OH, USA
| | - Ahmed Halima
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, R3 9500 Euclid Ave, Cleveland, 44195, OH, USA
| | - James B Broughman
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, R3 9500 Euclid Ave, Cleveland, 44195, OH, USA
| | - Chandana A Reddy
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, R3 9500 Euclid Ave, Cleveland, 44195, OH, USA
| | - Shireen Parsai
- Department of Radiation Oncology, Ohio Health Riverside Methodist Hospital, Columbus, OH, USA
| | - Jacob G Scott
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, R3 9500 Euclid Ave, Cleveland, 44195, OH, USA
| | - Timothy Chan
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, R3 9500 Euclid Ave, Cleveland, 44195, OH, USA
| | - Shauna Campbell
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, R3 9500 Euclid Ave, Cleveland, 44195, OH, USA
| | - Lilyana Angelov
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH, USA
| | - Stacey Zahler
- Department of Pediatric Hematology Oncology and Blood and Marrow Transplantation, Cleveland Clinic, Cleveland, OH, USA
| | - Matteo Trucco
- Department of Pediatric Hematology Oncology and Blood and Marrow Transplantation, Cleveland Clinic, Cleveland, OH, USA
| | - Stefanie M Thomas
- Department of Pediatric Hematology Oncology and Blood and Marrow Transplantation, Cleveland Clinic, Cleveland, OH, USA
| | - Shavaughn Johnson
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, R3 9500 Euclid Ave, Cleveland, 44195, OH, USA
| | - Peng Qi
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, R3 9500 Euclid Ave, Cleveland, 44195, OH, USA
| | - Anthony Magnelli
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, R3 9500 Euclid Ave, Cleveland, 44195, OH, USA
| | - Erin S Murphy
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, R3 9500 Euclid Ave, Cleveland, 44195, OH, USA.
| |
Collapse
|
2
|
Somasundaram E, Smile TD, Halima A, Broughman JB, Reddy CA, Parsai S, Scott JG, Chan T, Campbell S, Angelov L, Zahler S, Trucco M, Thomas SM, Johnson S, Qi P, Magnelli A, Anderson PM, Murphy ES. Neutrophil to Lymphocyte Ratio (NTLR) Predicts Local Control Failure and Overall Survival after Stereotactic Body Radiotherapy (SBRT) In Metastatic Sarcoma. RESEARCH SQUARE 2023:rs.3.rs-2570832. [PMID: 37333401 PMCID: PMC10275040 DOI: 10.21203/rs.3.rs-2570832/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
The neutrophil to lymphocyte ratio (NTLR) and absolute lymphocyte count (ALC) recovery are prognostic across many cancers. We investigated whether NLTR predicts SBRT success or survival in a metastatic sarcoma cohort treated with SBRT from 2014 and 2020 (N = 42). Wilcox Signed Rank Test and Friedman Test compare NTLR changes with local failure vs. local control (N = 138 lesions). Cox analyses identified factors associated with overall survival. If local control was successful, NLTR change was not significant (p = 0.30). However, NLTR significantly changed in patients local failure (p = 0.027). The multivariable Cox model demonstrated higher NLTR before SBRT was associated with worse overall survival (p = 0.002). The optimal NTLR cut point was 5 (Youden index: 0.418). One-year overall survival in SBRT metastatic sarcoma cohort was 47.6% (CI 34.3%-66.1%). Patients with an NTLR above 5 had a one-year overall survival of 37.7% (21.4%-66.3%); patients with an NTLR below 5 had a significantly improved overall survival of 63% (43.3%-91.6%, p = 0.014). Since NTLR at the time of SBRT was significantly associated with local control success and overall survival in metastatic sarcoma treated with SBRT, future efforts to reduce tumor inhibitory microenvironment factors and improved lymphocyte recovery should be investigated.
Collapse
|
3
|
Palombo R, Passacantilli I, Terracciano F, Capone A, Matteocci A, Tournier S, Alberdi A, Chiurchiù V, Volpe E, Paronetto MP. Inhibition of the PI3K/AKT/mTOR signaling promotes an M1 macrophage switch by repressing the ATF3-CXCL8 axis in Ewing sarcoma. Cancer Lett 2023; 555:216042. [PMID: 36565919 DOI: 10.1016/j.canlet.2022.216042] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/08/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
Ewing sarcomas are aggressive pediatric tumors of bone and soft tissues driven by in frame chromosomal translocations that yield fusion proteins guiding the oncogenic program. Promising alternative strategies to ameliorate current treatments involve inhibition of the PI3K/AKT/mTOR pathway. In this study, we identified the activating transcription factor 3 (ATF3) as an important mediator of the PI3K/AKT/mTOR pathway in Ewing sarcoma cells. ATF3 exerted its pro-tumoral activity through modulation of several chemokine-encoding genes, including CXCL8. The product of CXCL8, IL-8, acts as a pro-inflammatory chemokine critical for cancer progression and metastasis. We found that ATF3/IL-8 axis impacts macrophages populating the surrounding tumor microenvironment by promoting the M2 phenotype. Our study reveals valuable information on the PI3K/AKT/mTOR derived chemokine signaling in Ewing sarcoma cells: by promoting ATF3 and CXCL8 downregulation, inhibition of the PI3K/AKT/mTOR signaling promotes a proinflammatory response leading to upregulation of the protective anti-tumoral M1 macrophages.
Collapse
Affiliation(s)
- Ramona Palombo
- Laboratories of Molecular and Cellular Neurobiology, Molecular Neuroimmunology, and Resolution of Neuroinflammation, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano, 64, 00143, Rome, Italy; University of Rome "Foro Italico", Piazza Lauro de Bosis 6, 00135, Rome, Italy
| | - Ilaria Passacantilli
- Laboratories of Molecular and Cellular Neurobiology, Molecular Neuroimmunology, and Resolution of Neuroinflammation, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano, 64, 00143, Rome, Italy
| | - Francesca Terracciano
- Laboratories of Molecular and Cellular Neurobiology, Molecular Neuroimmunology, and Resolution of Neuroinflammation, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano, 64, 00143, Rome, Italy
| | - Alessia Capone
- Laboratories of Molecular and Cellular Neurobiology, Molecular Neuroimmunology, and Resolution of Neuroinflammation, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano, 64, 00143, Rome, Italy
| | - Alessandro Matteocci
- Laboratories of Molecular and Cellular Neurobiology, Molecular Neuroimmunology, and Resolution of Neuroinflammation, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano, 64, 00143, Rome, Italy
| | - Simon Tournier
- Plateforme Technologique IRSL UMS Saint-Louis US53 / UAR2030, Institut de Recherche Saint Louis, Université Paris Cité, France
| | - Antonio Alberdi
- Plateforme Technologique IRSL UMS Saint-Louis US53 / UAR2030, Institut de Recherche Saint Louis, Université Paris Cité, France
| | - Valerio Chiurchiù
- Laboratories of Molecular and Cellular Neurobiology, Molecular Neuroimmunology, and Resolution of Neuroinflammation, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano, 64, 00143, Rome, Italy; Institute of Translational Pharmacology, CNR, Via del Fosso del Cavaliere, 100, 00133, Rome, Italy
| | - Elisabetta Volpe
- Laboratories of Molecular and Cellular Neurobiology, Molecular Neuroimmunology, and Resolution of Neuroinflammation, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano, 64, 00143, Rome, Italy
| | - Maria Paola Paronetto
- Laboratories of Molecular and Cellular Neurobiology, Molecular Neuroimmunology, and Resolution of Neuroinflammation, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano, 64, 00143, Rome, Italy; University of Rome "Foro Italico", Piazza Lauro de Bosis 6, 00135, Rome, Italy.
| |
Collapse
|
4
|
Amin HM, Morani AC, Daw NC, Lamhamedi-Cherradi SE, Subbiah V, Menegaz BA, Vishwamitra D, Eskandari G, George B, Benjamin RS, Patel S, Song J, Lazar AJ, Wang WL, Kurzrock R, Pappo A, Anderson PM, Schwartz GK, Araujo D, Cuglievan B, Ratan R, McCall D, Mohiuddin S, Livingston JA, Molina ER, Naing A, Ludwig JA. IGF-1R/mTOR Targeted Therapy for Ewing Sarcoma: A Meta-Analysis of Five IGF-1R-Related Trials Matched to Proteomic and Radiologic Predictive Biomarkers. Cancers (Basel) 2020; 12:cancers12071768. [PMID: 32630797 PMCID: PMC7408058 DOI: 10.3390/cancers12071768] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 12/29/2022] Open
Abstract
Background : Ten to fourteen percent of Ewing sarcoma (ES) study participants treated nationwide with IGF-1 receptor (IGF-1R)-targeted antibodies achieved tumor regression. Despite this success, low response rates and short response durations (approximately 7-weeks) have slowed the development of this therapy. Methods: We performed a meta-analysis of five phase-1b/2 ES-oriented trials that evaluated the anticancer activity of IGF-1R antibodies +/− mTOR inhibitors (mTORi). Our meta-analysis provided a head-to-head comparison of the clinical benefits of IGF-1R antibodies vs. the IGF-1R/mTOR-targeted combination. Available pretreatment clinical samples were semi-quantitatively scored using immunohistochemistry to detect proteins in the IGF-1R/PI3K/AKT/mTOR pathway linked to clinical response. Early PET/CT imaging, obtained within the first 2 weeks (median 10 days), were examined to determine if reduced FDG avidity was predictive of progression-free survival (PFS). Results: Among 56 ES patients treated at MD Anderson Cancer Center (MDACC) with IGF-1R antibodies, our analysis revealed a significant ~two-fold improvement in PFS that favored a combination of IGF-1R/mTORi therapy (1.6 vs. 3.3-months, p = 0.042). Low pIGF-1R in the pretreatment specimens was associated with treatment response. Reduced total-lesion glycolysis more accurately predicted the IGF-1R response than other previously reported radiological biomarkers. Conclusion: Synergistic drug combinations, and newly identified proteomic or radiological biomarkers of IGF-1R response, may be incorporated into future IGF-1R-related trials to improve the response rate in ES patients.
Collapse
Affiliation(s)
- Hesham M. Amin
- Department of Hematopathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (H.M.A.); (D.V.); (G.E.); (B.G.)
| | - Ajaykumar C. Morani
- Department of Nuclear Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Najat C. Daw
- Department of Pediatrics, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (N.C.D.); (B.C.); (D.M.); (S.M.)
| | - Salah-Eddine Lamhamedi-Cherradi
- Department of Sarcoma Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (S.-E.L.-C.); (R.S.B.); (S.P.); (D.A.); (R.R.); (J.A.L.)
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics, 7Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (V.S.); (A.N.)
| | - Brian A. Menegaz
- Baylor College of Medicine, Department of Surgery, Breast Surgical Oncology, Houston, TX 77030, USA; (B.A.M.); (E.R.M.)
| | - Deeksha Vishwamitra
- Department of Hematopathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (H.M.A.); (D.V.); (G.E.); (B.G.)
| | - Ghazaleh Eskandari
- Department of Hematopathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (H.M.A.); (D.V.); (G.E.); (B.G.)
| | - Bhawana George
- Department of Hematopathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (H.M.A.); (D.V.); (G.E.); (B.G.)
| | - Robert S. Benjamin
- Department of Sarcoma Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (S.-E.L.-C.); (R.S.B.); (S.P.); (D.A.); (R.R.); (J.A.L.)
| | - Shreyaskumar Patel
- Department of Sarcoma Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (S.-E.L.-C.); (R.S.B.); (S.P.); (D.A.); (R.R.); (J.A.L.)
| | - Juhee Song
- Department of Biostatistics, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Alexander J. Lazar
- Department of Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.J.L.); (W.-L.W.)
| | - Wei-Lien Wang
- Department of Pathology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.J.L.); (W.-L.W.)
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy, University of California San Diego (UCSD) Moores Cancer Center, San Diego, CA 92037, USA;
| | - Alberto Pappo
- Department of Pathology, St. Jude’s Cancer Research Hospital, Memphis, TN 38105, USA;
| | | | - Gary K. Schwartz
- Division of Hematology & Oncology, Columbia University Medical Center, New York, NY 10032, USA;
| | - Dejka Araujo
- Department of Sarcoma Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (S.-E.L.-C.); (R.S.B.); (S.P.); (D.A.); (R.R.); (J.A.L.)
| | - Branko Cuglievan
- Department of Pediatrics, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (N.C.D.); (B.C.); (D.M.); (S.M.)
| | - Ravin Ratan
- Department of Sarcoma Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (S.-E.L.-C.); (R.S.B.); (S.P.); (D.A.); (R.R.); (J.A.L.)
| | - David McCall
- Department of Pediatrics, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (N.C.D.); (B.C.); (D.M.); (S.M.)
| | - Sana Mohiuddin
- Department of Pediatrics, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (N.C.D.); (B.C.); (D.M.); (S.M.)
| | - John A. Livingston
- Department of Sarcoma Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (S.-E.L.-C.); (R.S.B.); (S.P.); (D.A.); (R.R.); (J.A.L.)
| | - Eric R. Molina
- Baylor College of Medicine, Department of Surgery, Breast Surgical Oncology, Houston, TX 77030, USA; (B.A.M.); (E.R.M.)
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, 7Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (V.S.); (A.N.)
| | - Joseph A. Ludwig
- Department of Sarcoma Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (S.-E.L.-C.); (R.S.B.); (S.P.); (D.A.); (R.R.); (J.A.L.)
- Correspondence: ; Tel.: +1-(713)-792-3626
| |
Collapse
|
5
|
Heitzeneder S, Sotillo E, Shern JF, Sindiri S, Xu P, Jones R, Pollak M, Noer PR, Lorette J, Fazli L, Alag A, Meltzer P, Lau C, Conover CA, Oxvig C, Sorensen PH, Maris JM, Khan J, Mackall CL. Pregnancy-Associated Plasma Protein-A (PAPP-A) in Ewing Sarcoma: Role in Tumor Growth and Immune Evasion. J Natl Cancer Inst 2020; 111:970-982. [PMID: 30698726 DOI: 10.1093/jnci/djy209] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/10/2018] [Accepted: 11/12/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Ewing sarcoma (EWS) manifests one of the lowest somatic mutation rates of any cancer, leading to a scarcity of druggable mutations and neoantigens. Immunotherapeutics targeting differentially expressed cell surface antigens could provide therapeutic benefit for such tumors. Pregnancy-associated plasma protein A (PAPP-A) is a cell membrane-associated proteinase produced by the placenta that promotes fetal growth by inducing insulinlike growth factor (IGF) signaling. METHODS By comparing RNA expression of cell surface proteins in EWS (n = 120) versus normal tissues (n = 42), we comprehensively characterized the surfaceome of EWS to identify highly differentially expressed molecules. Using CRISPR/Cas-9 and anti-PAPP-A antibodies, we investigated biological roles for PAPP-A in EWS in vitro and in vivo in NSG xenograft models and performed RNA-sequencing on PAPPA knockout clones (n = 5) and controls (n = 3). All statistical tests were two-sided. RESULTS EWS surfaceome analysis identified 11 highly differentially overexpressed genes, with PAPPA ranking second in differential expression. In EWS cell lines, genetic knockout of PAPPA and treatment with anti-PAPP-A antibodies revealed an essential survival role by regulating local IGF-1 bioavailability. MAb-mediated PAPPA inhibition diminished EWS growth in orthotopic xenografts (leg area mm2 at day 49 IgG2a control (CTRL) [n = 14], mean = 397.0, SD = 86.1 vs anti-PAPP-A [n = 14], mean = 311.7, SD = 155.0; P = .03; median OS anti-PAPP-A = 52.5 days, 95% CI = 46.0 to 63.0 days vs IgG2a = 45.0 days, 95% CI = 42.0 to 52.0 days; P = .02) and improved the efficacy of anti-IGF-1R treatment (leg area mm2 at day 49 anti-PAPP-A + anti-IGF-1R [n = 15], mean = 217.9, SD = 148.5 vs IgG2a-CTRL; P < .001; median OS anti-PAPP-A + anti-IGF1R = 63.0 days, 95% CI = 52.0 to 67.0 days vs IgG2a-CTRL; P < .001). Unexpectedly, PAPPA knockout in EWS cell lines induced interferon (IFN)-response genes, including proteins associated with antigen processing/presentation. Consistently, gene expression profiles in PAPPA-low EWS tumors were enriched for immune response pathways. CONCLUSION This work provides a comprehensive characterization of the surfaceome of EWS, credentials PAPP-A as a highly differentially expressed therapeutic target, and discovers a novel link between IGF-1 signaling and immune evasion in cancer, thus implicating shared mechanisms of immune evasion between EWS and the placenta.
Collapse
|
6
|
Cesne AL, Bauer S, Demetri GD, Han G, Dezzani L, Ahmad Q, Blay JY, Judson I, Schöffski P, Aglietta M, Hohenberger P, Gelderblom H. Safety and efficacy of Pazopanib in advanced soft tissue sarcoma: PALETTE (EORTC 62072) subgroup analyses. BMC Cancer 2019; 19:794. [PMID: 31409302 PMCID: PMC6691522 DOI: 10.1186/s12885-019-5988-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 07/29/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND PALETTE is a phase 3 trial that demonstrated single-agent activity of pazopanib in advanced soft tissue sarcomas (aSTS). We performed retrospective subgroup analyses to explore potential relationships between patient characteristics, prior lines of therapy, dose intensity, and dose modifications on safety and efficacy of pazopanib in aSTS. METHODS PALETTE compared pazopanib with placebo in patients with aSTS (age ≥ 18 years) whose disease had progressed during or following prior chemotherapy. In these subgroup analyses, median progression-free survival (mPFS) among patients receiving pazopanib was the efficacy outcome of interest. Adverse events (AEs) were also compared within subgroups. All analyses were descriptive and exploratory. RESULTS A total of 246 patients received pazopanib in the PALETTE study. The mPFS was longer in patients who had only 1 prior line versus 2+ prior lines of therapy (24.7 vs 18.9 weeks, respectively); AE rates were similar regardless of number of prior lines of therapy. The mPFS was similar in patients aged < 65 and ≥ 65 y (20.0 and 20.1 weeks, respectively). Although AEs leading to study discontinuation were higher in older patients (≥65 y, 30%; < 65 y, 17%), rates of dose reductions, dose interruptions, and serious AEs were similar between the 2 age groups. No reduction in mPFS was noted in patients requiring dose reductions or dose interruptions to manage toxicities. CONCLUSIONS Longer mPFS was observed in patients receiving pazopanib following only 1 line of therapy. Additionally, mPFS with pazopanib was maintained regardless of patient age or dose modifications used to manage toxicity. TRIAL REGISTRATION NCT00753688 , first posted September 16, 2008 (registered prospectively).
Collapse
Affiliation(s)
- Axel Le Cesne
- Département d’Oncologie Médicale, Gustave Roussy, 114 Rue Edouard Vaillant, 94805 Villejuif Cedex, Villejuif, France
| | - Sebastian Bauer
- Sarcoma Center, West German Cancer Center, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany
| | - George D. Demetri
- Ludwig Center at Harvard, Harvard Medical School and Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215 USA
| | - Guangyang Han
- Novartis Oncology, One Health Plaza, East Hanover, NJ 07936 USA
| | - Luca Dezzani
- Novartis Oncology, One Health Plaza, East Hanover, NJ 07936 USA
| | - Qasim Ahmad
- Novartis Oncology, One Health Plaza, East Hanover, NJ 07936 USA
| | - Jean-Yves Blay
- Department of Medical Oncology, Leon Berard Center, 28, rue Laennec 2 69373 Lyon Cedex 08, Lyon, France
| | - Ian Judson
- The Institute of Cancer Research, Royal Marsden NHS Foundation Trust, 123 Old Brompton Road, London, SW7 3RP UK
| | - Patrick Schöffski
- Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Herestraat 49, B-3000 Leuven, Belgium
| | - Massimo Aglietta
- Department of Oncology, University of Torino and Candiolo Cancer Center FPO-IRCCS, 10060 Candiolo, (Torino) Italy
| | - Peter Hohenberger
- Division of Surgical Oncology & Thoracic Surgery, Mannheim University Medical Center, Theodor Kutzer Ufer 1, D-68165 Mannheim, Germany
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, 2333 Leiden, ZA Netherlands
| |
Collapse
|
7
|
Pei J, Zhao X, Patchefsky AS, Flieder DB, Talarchek JN, Testa JR, Wei S. Clinical application of RNA sequencing in sarcoma diagnosis: An institutional experience. Medicine (Baltimore) 2019; 98:e16031. [PMID: 31232935 PMCID: PMC6636967 DOI: 10.1097/md.0000000000016031] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Accurate diagnoses of sarcoma are sometimes challenging on conventional histomorphology and immunophenotype. Many specific genetic aberrations including chromosomal translocations have been identified in various sarcomas, which can be detected by fluorescence in situ hybridization and polymerase chain reaction analysis. Next-generation sequencing-based RNA sequencing can screen multiple sarcoma-specific chromosome translocations/fusion genes in 1 test, which is especially useful for sarcoma without obvious differentiation. In this report, we utilized RNA sequencing on formalin-fixed paraffin-embedded (FFPE) specimens to investigate the possibility of diagnosing sarcomas by identifying disease-specific fusion genes. Targeted RNA sequencing was performed on 6 sarcoma cases. The expected genetic alterations (clear cell sarcoma/EWSR1-ATF1, Ewing sarcoma/EWSR1-FLI1, myxoid liposarcoma/DDIT3-FUS) in four cases were detected and confirmed by secondary tests. Interestingly, three SS18 fusion genes (SS18-SSX2B, SS18-SSX2, and SS18-SSX4) were identified in a synovial sarcoma case. A rare fusion gene (EWSR1-PATZ1) was identified in a morphologically challenging case; which enabled us to establish the diagnosis of low grade glioneural tumor. In conclusion, RNA sequencing on FFPE specimen is a reliable method in establishing the diagnosis of sarcoma in daily practice.
Collapse
Affiliation(s)
| | - Xiaofeng Zhao
- Department of Pathology and Laboratory Medicine, Temple University Hospital
| | | | | | | | - Joseph R. Testa
- Genomics Facility, Fox Chase Cancer Center
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA
| | | |
Collapse
|
8
|
Groisberg R, Hong DS, Holla V, Janku F, Piha-Paul S, Ravi V, Benjamin R, Kumar Patel S, Somaiah N, Conley A, Ali SM, Schrock AB, Ross JS, Stephens PJ, Miller VA, Sen S, Herzog C, Meric-Bernstam F, Subbiah V. Clinical genomic profiling to identify actionable alterations for investigational therapies in patients with diverse sarcomas. Oncotarget 2018; 8:39254-39267. [PMID: 28424409 PMCID: PMC5503611 DOI: 10.18632/oncotarget.16845] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 03/08/2017] [Indexed: 12/22/2022] Open
Abstract
Background There are currently no United States Food and Drug Administration approved molecularly matched therapies for sarcomas except gastrointestinal stromal tumors. Complicating this is the extreme diversity, heterogeneity, and rarity of these neoplasms. Few therapeutic options exist for relapsed and refractory sarcomas. In clinical practice many oncologists refer patients for genomic profiling hoping for guidance on treatment options after standard therapy. However, a systematic analysis of actionable mutations has yet to be completed. We analyzed genomic profiling results in patients referred to MD Anderson Cancer Center with advanced sarcomas to elucidate the frequency of potentially actionable genomic alterations in this population. Methods We reviewed charts of patients with advanced sarcoma who were referred to investigational cancer therapeutics department and had CLIA certified comprehensive genomic profiling (CGP) of 236 or 315 cancer genes in at least 50ng of DNA. Actionable alterations were defined as those identifying anti-cancer drugs on the market, in registered clinical trials, or in the Drug-Gene Interaction Database. Results Among the 102 patients analyzed median age was 45.5 years (range 8-76), M: F ratio 48:54. The most common subtypes seen in our study were leiomyosarcoma (18.6%), dedifferentiated liposarcoma (11%), osteosarcoma (11%), well-differentiated liposarcoma (7%), carcinosarcoma (6%), and rhabdomyosarcoma (6%). Ninety-five out of 102 patients (93%) had at least one genomic alteration identified with a mean of six mutations per patient. Of the 95 biopsy samples with identifiable genomic alterations, the most commonly affected genes were TP53 (31.4%), CDK4 (23.5%), MDM2 (21.6%), RB1 (18.6%), and CDKN2A/B (13.7%). Notable co-segregating amplifications included MDM2-CDK4 and FRS2-FGF. Sixteen percent of patients received targeted therapy based on CGP of which 50% had at least stable disease. Conclusions Incorporating CGP into sarcoma management may allow for more precise diagnosis and sub-classification of this diverse and rare disease, as well as personalized matching of patients to targeted therapies such as those available in basket clinical trials.
Collapse
Affiliation(s)
- Roman Groisberg
- Department of Investigational Cancer Therapeutics (A Phase I Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.,Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics (A Phase I Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Vijaykumar Holla
- Khalifa Institute for Personalized Cancer Therapy (IPCT), The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Filip Janku
- Department of Investigational Cancer Therapeutics (A Phase I Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Sarina Piha-Paul
- Department of Investigational Cancer Therapeutics (A Phase I Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Vinod Ravi
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Robert Benjamin
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Shreyas Kumar Patel
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Neeta Somaiah
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Anthony Conley
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Siraj M Ali
- Foundation Medicine Inc, Cambridge, Massachusetts 02139, USA
| | - Alexa B Schrock
- Foundation Medicine Inc, Cambridge, Massachusetts 02139, USA
| | - Jeffrey S Ross
- Foundation Medicine Inc, Cambridge, Massachusetts 02139, USA
| | | | | | - Shiraj Sen
- Department of Investigational Cancer Therapeutics (A Phase I Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.,Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Cynthia Herzog
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics (A Phase I Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics (A Phase I Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
9
|
Groisberg R, Hong DS, Behrang A, Hess K, Janku F, Piha-Paul S, Naing A, Fu S, Benjamin R, Patel S, Somaiah N, Conley A, Meric-Bernstam F, Subbiah V. Characteristics and outcomes of patients with advanced sarcoma enrolled in early phase immunotherapy trials. J Immunother Cancer 2017; 5:100. [PMID: 29254498 PMCID: PMC5735899 DOI: 10.1186/s40425-017-0301-y] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 11/08/2017] [Indexed: 12/16/2022] Open
Abstract
Background Immunotherapies, specifically those based on immune checkpoint inhibitors, have shown promising activity in multiple tumor types. Other than mifamurtide (MEPACT®) for osteosarcoma approved by European Medicines Agency, there are no approved immunotherapies for sarcomas. Methods We analyzed medical records of patients with advanced sarcoma who were referred to Phase 1 clinic at MD Anderson and received an immunotherapy (checkpoint inhibitors, vaccines, or cytokine based therapies). Clinical parameters including demographics, clinical history, toxicity, and response were abstracted. Results Among 50 patients enrolled in immunotherapy trials (Bone 10; Soft-tissue 40) we found 14 different subtypes of sarcomas. Royal Marsden Hospital (RMH) prognostic score was <2 (86%). Performance status (PS) was 0–1 in 48 patients (96%); median number of prior therapies was 3 (0–12). Immunotherapy consisted of checkpoint inhibitors (82%: PD1 = 7, PD-L1 = 11, CTLA4 = 22, other = 1) of which 42% were combinations, as well as vaccines (14%), and cytokines (4%). Median overall survival (OS) was 13.4 months (11.2 months: not reached). Median progression free survival (PFS) was 2.4 months (95% CI = 1.9–3.2 months). Best response was partial response (PR) in 2 patients with alveolar soft part sarcoma (ASPS) and stable disease (SD) in 11 patients (3 GIST, 3 liposarcomas (2 DDLS, 1 WDLS), 2 ASPS, 2 leiomyo, 1 osteo). PFS was 34% (23%, at 50%) at 3 months, 16% (8%, 30%) at 6 months, and 6% (2%, 20%) at 1 year. Pseudo-progression followed by stable disease was observed in 2 patients (4%). Grade 3/4 adverse events included rash (10%), fever (6%), fatigue (6%), and nausea/vomiting (6%). Conclusion Immunotherapies were well tolerated in advanced sarcoma patients enrolled in trials. All four ASPS patients had clinical benefit with checkpoint inhibitors and this was the only subtype experiencing partial response. Further evaluation of checkpoint inhibitors in ASPS is warranted.
Collapse
Affiliation(s)
- Roman Groisberg
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Unit 455, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.,Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Unit 455, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Amini Behrang
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kenneth Hess
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Filip Janku
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Unit 455, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Sarina Piha-Paul
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Unit 455, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Aung Naing
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Unit 455, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Siqing Fu
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Unit 455, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Robert Benjamin
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Shreyaskumar Patel
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Neeta Somaiah
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Anthony Conley
- Department of Sarcoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Unit 455, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Unit 455, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| |
Collapse
|
10
|
Outcomes of patients with sarcoma enrolled in clinical trials of pazopanib combined with histone deacetylase, mTOR, Her2, or MEK inhibitors. Sci Rep 2017; 7:15963. [PMID: 29162825 PMCID: PMC5698336 DOI: 10.1038/s41598-017-13114-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 09/19/2017] [Indexed: 12/29/2022] Open
Abstract
Pazopanib is US FDA approved for the treatment of advanced soft tissue sarcomas. All patients with this disease ultimately develop resistance to therapy. Mechanisms of resistance include activation of the mTOR, histone deacetylase (HDAC), MAPK, and ERBB4 pathways. We hypothesized that combining pazopanib with other targeted agents inhibiting these pathways would increase response rates. We retrospectively evaluated the safety and efficacy of pazopanib plus vorinostat, everolimus, lapatinib or trastuzumab, and MEK inhibitor in patients with advanced sarcoma. The Cancer Geneome Atlas (TCGA) data was analyzed for HDAC, PI3K, HER2, and MAPK/RAS/RAF gene alterations from sarcoma TCGA. Of the 44 advanced sarcoma patients in these trials, 27 (61%) were male; 18 (41%) had bone sarcoma, and 26 (59%) had soft tissue sarcoma. Best response was partial response (PR) in four patients [(overall response rate (ORR) = 9%, 95% confidence interval [CI] 3% to 22%)]. The median progression-free survival (PFS) for all patients was 9.6 weeks (95% CI 8.0 to 15.7 weeks). Analysis of TCGA data revealed HDAC, PI3K, HER2, and MAPK/RAS/RAF gene alterations in 112/243 (46%) of patients predominantly HDAC1-11 (41%) alterations. Pazopanib combinations did demonstrate safety in combination with other agents. TCGA data suggests further evaluation of epigenetic pathway inhibitors in sarcoma.
Collapse
|
11
|
The Role of Next-Generation Sequencing in Sarcomas: Evolution From Light Microscope to Molecular Microscope. Curr Oncol Rep 2017; 19:78. [DOI: 10.1007/s11912-017-0641-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|