1
|
Lee AQ, Hao C, Pan M, Ganjoo KN, Bui NQ. Histologic and Immunologic Factors Associated with Response to Immune Checkpoint Inhibitors in Advanced Sarcoma. Clin Cancer Res 2025; 31:678-684. [PMID: 39699310 DOI: 10.1158/1078-0432.ccr-24-3485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/10/2024] [Accepted: 12/16/2024] [Indexed: 12/20/2024]
Abstract
PURPOSE To characterize factors associated with response to immune checkpoint inhibitors (ICI) in advanced sarcoma. EXPERIMENTAL DESIGN This is a retrospective study with a cohort of 216 patients with advanced sarcoma treated with ICI between 2016 and 2023 at Stanford Health Care. Overall survival, progression-free survival (PFS), objective response rates (ORR) per RECIST criteria, and reason for ICI discontinuation were analyzed across histologic subtypes, ICI regimens, tumor mutational burden, and PD-L1 expression. RESULTS The overall ORR in the cohort was 16.7%. The histologic subtypes with the highest ORR were Kaposi sarcoma (KS, 66.7%), alveolar soft part sarcoma (ASPS, 50%), angiosarcoma (33.3%), myxofibrosarcoma (MFS, 28.6%), and undifferentiated pleomorphic sarcoma (UPS, 27.8%). The subtypes with the lowest ORR were osteosarcoma (0%), synovial sarcoma (0%), and liposarcoma (3.7%). The subtypes with the highest median PFS were KS (median not reached), ASPS (median not reached), MFS (27.4 months), and UPS (11.3 months). The ORR for sarcomas with PD-L1 ≥ 1% was 27.8% (P = 0.02), whereas the ORR for sarcomas with tumor mutational burden ≥10 mutations per megabase of DNA was 28.6% (P = 0.20). CONCLUSIONS ORR and PFS were highly variable across sarcoma histologic subtypes. In this large analysis, KS, ASPS, angiosarcoma, MFS, and UPS demonstrated the highest ORR and longest PFS while osteosarcoma, synovial sarcoma, and liposarcoma had the lowest ORR and shortest PFS. PD-L1 expression was also associated with increased ORR. Our findings provide further insight into understanding the sarcoma histologic and immunologic factors that correspond with response to ICI.
Collapse
Affiliation(s)
- Alex Q Lee
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Clara Hao
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Minggui Pan
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Kristen N Ganjoo
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Nam Q Bui
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
2
|
Hebron KE, Perkins OL, Kim A, Jian X, Girald-Berlingeri SA, Lei H, Shern JF, Conner EA, Randazzo PA, Yohe ME. ASAP1 and ARF1 Regulate Myogenic Differentiation in Rhabdomyosarcoma by Modulating TAZ Activity. Mol Cancer Res 2025; 23:95-106. [PMID: 39495123 PMCID: PMC11799837 DOI: 10.1158/1541-7786.mcr-24-0490] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/08/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Despite aggressive, multimodal therapies, the prognosis of patients with refractory or recurrent rhabdomyosarcoma (RMS) has not improved in four decades. Because RMS resembles skeletal muscle precursor cells, differentiation-inducing therapy has been proposed for patients with advanced disease. In RAS-mutant PAX fusion-negative RMS (FN-RMS) preclinical models, MEK1/2 inhibition (MEKi) induces differentiation, slows tumor growth, and extends survival. However, the response is short-lived. A better understanding of the molecular mechanisms regulating FN-RMS differentiation could improve differentiation therapy. In this study, we identified a role in FN-RMS differentiation for ASAP1, an ADP ribosylation factor (ARF) GTPase-activating protein (GAP) with both proinvasive and tumor-suppressor functions. We found that ASAP1 knockdown inhibited differentiation in FN-RMS cells. Interestingly, knockdown of the GTPases ARF1 or ARF5, targets of ASAP1 GAP activity, also blocked differentiation of FN-RMS. We discovered that loss of ARF pathway components blocked myogenic transcription factor expression. Therefore, we examined the effects on transcriptional regulators. MEKi led to the phosphorylation and inactivation of WW domain-containing transcriptional regulator 1 (WWTR1; TAZ), a homolog of the pro-proliferative transcriptional co-activator YAP1, regulated by the Hippo pathway. However, loss of ASAP1 or ARF1 blocked this inactivation, which inhibits MEKi-induced differentiation. Finally, MEKi-induced differentiation was rescued by dual knockdown of ASAP1 and WWTR1. This study shows that ASAP1 and ARF1 are necessary for myogenic differentiation, providing a deeper understanding of differentiation in FN-RMS and illuminating an opportunity to advance differentiation therapy. Implications: ASAP1 and ARF1 regulate MEKi-induced differentiation of FN-RMS cells by modulating WWTR1 (TAZ) activity, supporting YAP1/TAZ inhibition as a FN-RMS differentiation therapy strategy.
Collapse
Affiliation(s)
- Katie E. Hebron
- Laboratory of Cell and Developmental Signaling, National Cancer Institute, National Institutes of Health, Frederick, Maryland
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Laboratory of Cell and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Olivia L. Perkins
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Laboratory of Cell and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| | - Angela Kim
- Laboratory of Cell and Developmental Signaling, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | - Xiaoying Jian
- Laboratory of Cell and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sofia A. Girald-Berlingeri
- Laboratory of Cell and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Haiyan Lei
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jack F. Shern
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Elizabeth A. Conner
- Center for Cancer Research Genomics Core, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Paul A. Randazzo
- Laboratory of Cell and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Marielle E. Yohe
- Laboratory of Cell and Developmental Signaling, National Cancer Institute, National Institutes of Health, Frederick, Maryland
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Laboratory of Cell and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
3
|
Kuo C, Giannikou K, Wang N, Warren M, Goodspeed A, Shillingford N, Hayashi M, Raredon MSB, Amatruda JF. Tumor-associated stroma shapes the spatial tumor immune microenvironment of primary Ewing sarcomas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.31.635996. [PMID: 39975230 PMCID: PMC11838416 DOI: 10.1101/2025.01.31.635996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
To date, few studies have detailed the tumor microenvironment (TME) of Ewing sarcoma (EwS). The TME has a vital role in cancer survival and progression with implications in drug resistance and immune escape. By performing spatially resolved transcriptomic analysis of primary treatment-naïve EwS samples, we discovered greater stromal enrichment in localized EwS tumors compared to metastatic EwS tumors. Through spatial ligand-receptor analysis, we show that the stromal enriched regions harbor unique extracellular matrix related cytokines, immune recruitment and proinflammatory microenvironmental signals, implying EwS stroma may play an anti-tumorigenic role by acting as an immune recruitment center. All EwS tumors expressed pro-tumorigenic MIF-CD74 immune signaling, suggesting a potential immune-evasive mechanism and immunotherapy target. Our findings provide insight into tumor cell/stromal cell interactions in EwS and serve as a valuable resource for further investigations in the tumor immune microenvironment of EwS.
Collapse
Affiliation(s)
- Christopher Kuo
- Cancer and Blood Disease Institute, Division of Hematology-Oncology, Children's Hospital Los Angeles
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Krinio Giannikou
- Cancer and Blood Disease Institute, Division of Hematology-Oncology, Children's Hospital Los Angeles
| | - Nuoya Wang
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT
| | - Mikako Warren
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA
| | - Andrew Goodspeed
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO
- Department of Biomedical informatics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Nick Shillingford
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA
| | - Masanori Hayashi
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Micha Sam Brickman Raredon
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT
| | - James F Amatruda
- Cancer and Blood Disease Institute, Division of Hematology-Oncology, Children's Hospital Los Angeles
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA
| |
Collapse
|
4
|
Metts J, Rodriguez-Valentin M, Hensel J, Alfaro A, Snyder CW, Binitie O, Chebli C, Monforte H, Pilon-Thomas S, Mullinax J. Expansion of tumor-infiltrating and marrow-infiltrating lymphocytes from pediatric malignant solid tumors. Cytotherapy 2025; 27:29-35. [PMID: 39243253 PMCID: PMC11668621 DOI: 10.1016/j.jcyt.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/26/2024] [Accepted: 08/05/2024] [Indexed: 09/09/2024]
Abstract
INTRODUCTION The expansion of tumor-infiltrating lymphocytes (TIL) for adoptive cellular therapy is under investigation in many solid tumors of adulthood. Marrow-infiltrating lymphocytes (MIL) have demonstrated antitumor reactivity preclinically. Successful expansion of TIL/MIL has not been reported across pediatric solid tumor histologies. The objective of this study was to demonstrate successful expansion of TIL from pediatric solid tumors for translation in an adoptive cell therapy (ACT) treatment strategy. METHODS A prospective study of TIL/MIL expansion was performed on solid tumors of pediatric patients undergoing standard-of-care procedures. TIL/MIL expansions were performed in the presence of high-dose interleukin 2. To demonstrate a full-scale expansion to clinically-relevant cell doses for TIL therapy, initial TIL culture was followed by a rapid expansion protocol for select patients. Expanded specimens were analyzed for phenotype by flow cytometry and for anti-tumor reactivity by the interferon-gamma release assay. RESULTS Eighteen tumor samples were obtained. Initial TIL cultures were successfully generated from 14/18 samples (77.7%). A median of 5.52 × 107 (range: 2.5 × 106-3.23 × 108) cells were produced from initial cultures, with 46.9% expressing a CD3 phenotype (46.9%). Eight samples underwent rapid expansion, demonstrating a median 458-fold expansion and a CD3 phenotype of 98%. Initial MIL cultures were successfully generated from five samples, with a predominantly CD3 phenotype (45.2%). Sufficient tumor tissue was only available for seven TIL samples to be tested for reactivity; none demonstrated responsiveness to autologous tumor. CONCLUSIONS TIL and MIL expansion from pediatric solid tumors was successful, including the full-scale expansion process. This data supports translation to an ACT-TIL treatment strategy in the pediatric population and thus a Phase I trial of ACT-TIL in pediatric high-risk solid tumors is planned.
Collapse
Affiliation(s)
- Jonathan Metts
- Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St Petersburg, Florida, USA; Departments of Sarcoma, Immunology, and Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA.
| | | | | | - Alex Alfaro
- Departments of Sarcoma, Immunology, and Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Christopher W Snyder
- Division of Pediatric Surgery, Johns Hopkins All Children's Hospital, St Petersburg, Florida, USA
| | - Odion Binitie
- Departments of Sarcoma, Immunology, and Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Caroline Chebli
- Department of Orthopedic Surgery, James A Haley Veteran's Administration Hospital, Tampa, Florida, USA
| | - Hector Monforte
- Section of Anatomic Pathology, Johns Hopkins All Children's Hospital, St Petersburg, Florida, USA
| | - Shari Pilon-Thomas
- Departments of Sarcoma, Immunology, and Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - John Mullinax
- Departments of Sarcoma, Immunology, and Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| |
Collapse
|
5
|
Luo Y, Shentu J, Xu H, Xia Y, Fang L, Duan S. Innovative pan-tumor target strategy for CAR-T therapy: cancer-specific exons as novel targets for pediatric solid and brain tumors. J Transl Med 2024; 22:1019. [PMID: 39533264 PMCID: PMC11556178 DOI: 10.1186/s12967-024-05861-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) immunotherapy has achieved remarkable success in treating chemotherapy-refractory hematological malignancies. However, its efficacy in solid and brain tumors remains limited due to challenges such as insufficient target antigens, poor T-cell adaptability, inefficient tumor site trafficking, and the immunosuppressive tumor microenvironment. To address these challenges, Shaw and colleagues proposed an innovative strategy targeting cancer-specific exons (CSEs) in pediatric solid and brain tumors. Using RNA sequencing data from 16 tumor types, the study identified 157 highly tumor-specific targets, including both known and novel proteins. The researchers validated several targets, including FN1 and COL11A1, demonstrating their therapeutic potential in in vitro and in vivo models. The study's approach of integrating exon-level analysis with a broad search for extracellular matrix proteins offers a new frontier for CAR-T therapy, providing valuable insights for improving immunotherapy in pediatric solid tumors. Although promising, the study also highlights the need for further evaluation of tumor recurrence and CAR-T cell exhaustion. The identification of novel pan-tumor targets may revolutionize CAR-T therapy design and expand its application in pediatric cancer treatment.
Collapse
Affiliation(s)
- Yuqing Luo
- Department of Hematology, Yuyao People's Hospital of Zhejiang Province, The Affiliated Yangming Hospital of Ningbo University, Yuyao, 315400, Zhejiang, China
| | - Jianqiao Shentu
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang, China
| | - Hening Xu
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang, China
| | - Yongming Xia
- Department of Hematology, Yuyao People's Hospital of Zhejiang Province, The Affiliated Yangming Hospital of Ningbo University, Yuyao, 315400, Zhejiang, China
| | - Lili Fang
- Department of Hematology, Yuyao People's Hospital of Zhejiang Province, The Affiliated Yangming Hospital of Ningbo University, Yuyao, 315400, Zhejiang, China.
| | - Shiwei Duan
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang, China.
| |
Collapse
|
6
|
Morel VJ, Rössler J, Bernasconi M. Targeted immunotherapy and nanomedicine for rhabdomyosarcoma: The way of the future. Med Res Rev 2024; 44:2730-2773. [PMID: 38885148 DOI: 10.1002/med.22059] [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: 06/29/2023] [Revised: 04/17/2024] [Accepted: 05/20/2024] [Indexed: 06/20/2024]
Abstract
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma of childhood. Histology separates two main subtypes: embryonal RMS (eRMS; 60%-70%) and alveolar RMS (aRMS; 20%-30%). The aggressive aRMS carry one of two characteristic chromosomal translocations that result in the expression of a PAX3::FOXO1 or PAX7::FOXO1 fusion transcription factor; therefore, aRMS are now classified as fusion-positive (FP) RMS. Embryonal RMS have a better prognosis and are clinically indistinguishable from fusion-negative (FN) RMS. Next to histology and molecular characteristics, RMS risk groupings are now available defining low risk tumors with excellent outcomes and advanced stage disease with poor prognosis, with an overall survival of about only 20% despite intensified multimodal treatment. Therefore, development of novel effective targeted strategies to increase survival and to decrease long-term side effects is urgently needed. Recently, immunotherapies and nanomedicine have been emerging for potent and effective tumor treatments with minimal side effects, raising hopes for effective and safe cures for RMS patients. This review aims to describe the most relevant preclinical and clinical studies in immunotherapy and targeted nanomedicine performed so far in RMS and to provide an insight in future developments.
Collapse
Affiliation(s)
- Victoria Judith Morel
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Jochen Rössler
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Michele Bernasconi
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| |
Collapse
|
7
|
Robinson SI, Rochell RE, Penza V, Naik S. Translation of oncolytic viruses in sarcoma. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200822. [PMID: 39040851 PMCID: PMC11261849 DOI: 10.1016/j.omton.2024.200822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Sarcomas are a rare and highly diverse group of malignancies of mesenchymal origin. While sarcomas are generally considered resistant to immunotherapy, recent studies indicate subtype-specific differences in clinical response to checkpoint inhibitors (CPIs) that are associated with distinct immune phenotypes present in sarcoma subtypes. Oncolytic viruses (OVs) are designed to selectively infect and kill tumor cells and induce intratumoral immune infiltration, enhancing immunogenicity and thereby sensitizing tumors to immunotherapy. Herein we review the accumulated clinical data evaluating OVs in sarcoma. Small numbers of patients with sarcoma were enrolled in early-stage OV trials as part of larger solid tumor cohorts demonstrating safety but providing limited insight into the biological effects due to the low patient numbers and lack of histologic grouping. Several recent studies have investigated talimogene laherparepvec (T-VEC), an approved oncolytic herpes simplex virus (HSV-1), in combination therapy regimens in sarcoma patient cohorts. These studies have shown promising responses in heavily pre-treated and immunotherapy-resistant patients associated with increased intratumoral immune infiltration. As new and more potent OVs enter the clinical arena, prospective evaluation in subtype-specific cohorts with correlative studies to define biomarkers of response will be critical to advancing this promising approach for sarcoma therapy.
Collapse
Affiliation(s)
- Steven I. Robinson
- Division of Medical Oncology, Mayo Clinic, Rochester, MN 55902, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Roya E. Rochell
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Velia Penza
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Shruthi Naik
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
8
|
Petrescu DI, Yustein JT, Dasgupta A. Preclinical models for the study of pediatric solid tumors: focus on bone sarcomas. Front Oncol 2024; 14:1388484. [PMID: 39091911 PMCID: PMC11291195 DOI: 10.3389/fonc.2024.1388484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/01/2024] [Indexed: 08/04/2024] Open
Abstract
Sarcomas comprise between 10-15% of all pediatric malignancies. Osteosarcoma and Ewing sarcoma are the two most common pediatric bone tumors diagnosed in children and young adults. These tumors are commonly treated with surgery and/or radiation therapy and combination chemotherapy. However, there is a strong need for the development and utilization of targeted therapeutic methods to improve patient outcomes. Towards accomplishing this goal, pre-clinical models for these unique malignancies are of particular importance to design and test experimental therapeutic strategies prior to being introduced to patients due to their origination site and propensity to metastasize. Pre-clinical models offer several advantages for the study of pediatric sarcomas with unique benefits and shortcomings dependent on the type of model. This review addresses the types of pre-clinical models available for the study of pediatric solid tumors, with special attention to the bone sarcomas osteosarcoma and Ewing sarcoma.
Collapse
Affiliation(s)
- D. Isabel Petrescu
- Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, GA, United States
| | - Jason T. Yustein
- Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, GA, United States
| | - Atreyi Dasgupta
- The Faris D. Virani Ewing Sarcoma Center, Baylor College of Medicine, Texas Children’s Cancer and Hematology Centers, Houston, TX, United States
| |
Collapse
|
9
|
Sicinska E, Sudhakara Rao Kola V, Kerfoot JA, Taddei ML, Al-Ibraheemi A, Hsieh YH, Church AJ, Landesman-Bollag E, Landesman Y, Hemming ML. ASPSCR1::TFE3 Drives Alveolar Soft Part Sarcoma by Inducing Targetable Transcriptional Programs. Cancer Res 2024; 84:2247-2264. [PMID: 38657118 PMCID: PMC11250573 DOI: 10.1158/0008-5472.can-23-2115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 02/09/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024]
Abstract
Alveolar soft part sarcoma (ASPS) is a rare mesenchymal malignancy driven by the ASPSCR1::TFE3 fusion. A better understanding of the mechanisms by which this oncogenic transcriptional regulator drives cancer growth is needed to help identify potential therapeutic targets. In this study, we characterized the transcriptional and chromatin landscapes of ASPS tumors and preclinical models, identifying the essential role of ASPSCR1::TFE3 in tumor cell viability by regulating core transcriptional programs involved in cell proliferation, angiogenesis, and mitochondrial biology. ASPSCR1::TFE3 directly interacted with key epigenetic regulators at enhancers and promoters to support ASPS-associated transcription. Among the effector programs driven by ASPSCR1::TFE3, cell proliferation was driven by high levels of cyclin D1 expression. Disruption of cyclin D1/CDK4 signaling led to a loss of ASPS proliferative capacity, and combined inhibition of CDK4/6 and angiogenesis halted tumor growth in xenografts. These results define the ASPS oncogenic program, reveal mechanisms by which ASPSCR1::TFE3 controls tumor biology, and identify a strategy for therapeutically targeting tumor cell-intrinsic vulnerabilities. Significance: The ASPSCR1::TFE3 fusion propels the growth of alveolar soft part sarcoma by activating transcriptional programs that regulate proliferation, angiogenesis, mitochondrial biogenesis, and differentiation and can be therapeutically targeted to improve treatment.
Collapse
MESH Headings
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics
- Sarcoma, Alveolar Soft Part/genetics
- Sarcoma, Alveolar Soft Part/pathology
- Sarcoma, Alveolar Soft Part/metabolism
- Humans
- Animals
- Mice
- Cell Proliferation/genetics
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Gene Expression Regulation, Neoplastic
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
- Cyclin-Dependent Kinase 4/genetics
- Cyclin-Dependent Kinase 4/metabolism
- Cyclin-Dependent Kinase 4/antagonists & inhibitors
- Female
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/metabolism
- Intracellular Signaling Peptides and Proteins
Collapse
Affiliation(s)
- Ewa Sicinska
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Vijaya Sudhakara Rao Kola
- Department of Medicine, Division of Hematology and Oncology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Joseph A. Kerfoot
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Madeleine L. Taddei
- Department of Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Alyaa Al-Ibraheemi
- Department of Pathology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yi-Hsuan Hsieh
- Department of Medicine, Division of Hematology and Oncology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Alanna J. Church
- Department of Pathology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Esther Landesman-Bollag
- Department of Medicine, Section of Hematology and Oncology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Yosef Landesman
- Cure Alveolar Soft Part Sarcoma International, Brookline, Massachusetts, USA
| | - Matthew L. Hemming
- Department of Medicine, Division of Hematology and Oncology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
10
|
Ebegboni VJ, Jones TL, Brownmiller T, Zhao PX, Pehrsson EC, Rajan SS, Caplen NJ. ETS1, a Target Gene of the EWSR1::FLI1 Fusion Oncoprotein, Regulates the Expression of the Focal Adhesion Protein TENSIN3. Mol Cancer Res 2024; 22:625-641. [PMID: 38588446 PMCID: PMC11219265 DOI: 10.1158/1541-7786.mcr-23-1090] [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: 12/29/2023] [Revised: 03/25/2024] [Accepted: 04/08/2024] [Indexed: 04/10/2024]
Abstract
The mechanistic basis for the metastasis of Ewing sarcomas remains poorly understood, as these tumors harbor few mutations beyond the chromosomal translocation that initiates the disease. Instead, the epigenome of Ewing sarcoma cells reflects the regulatory state of genes associated with the DNA-binding activity of the fusion oncoproteins EWSR1::FLI1 or EWSR1::ERG. In this study, we examined the EWSR1::FLI1/ERG's repression of transcription factor genes, concentrating on those that exhibit a broader range of expression in tumors than in Ewing sarcoma cell lines. Focusing on one of these target genes, ETS1, we detected EWSR1::FLI1 binding and an H3K27me3-repressive mark at this locus. Depletion of EWSR1::FLI1 results in ETS1's binding of promoter regions, substantially altering the transcriptome of Ewing sarcoma cells, including the upregulation of the gene encoding TENSIN3 (TNS3), a focal adhesion protein. Ewing sarcoma cell lines expressing ETS1 (CRISPRa) exhibited increased TNS3 expression and enhanced movement compared with control cells. Visualization of control Ewing sarcoma cells showed a distributed vinculin signal and a network-like organization of F-actin; in contrast, ETS1-activated Ewing sarcoma cells showed an accumulation of vinculin and F-actin toward the plasma membrane. Interestingly, the phenotype of ETS1-activated Ewing sarcoma cell lines depleted of TNS3 resembled the phenotype of the control cells. Critically, these findings have clinical relevance as TNS3 expression in Ewing sarcoma tumors positively correlates with that of ETS1. Implications: ETS1's transcriptional regulation of the gene encoding the focal adhesion protein TENSIN3 in Ewing sarcoma cells promotes cell movement, a critical step in the evolution of metastasis.
Collapse
Affiliation(s)
- Vernon Justice Ebegboni
- Functional Genetics Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tamara L. Jones
- Functional Genetics Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tayvia Brownmiller
- Functional Genetics Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Patrick X. Zhao
- Omics Bioinformatics Facility, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Erica C. Pehrsson
- Omics Bioinformatics Facility, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Soumya Sundara Rajan
- Functional Genetics Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Natasha J. Caplen
- Functional Genetics Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
11
|
Hegde M, Navai S, DeRenzo C, Joseph SK, Sanber K, Wu M, Gad AZ, Janeway KA, Campbell M, Mullikin D, Nawas Z, Robertson C, Mathew PR, Zhang H, Mehta B, Bhat RR, Major A, Shree A, Gerken C, Kalra M, Chakraborty R, Thakkar SG, Dakhova O, Salsman VS, Grilley B, Lapteva N, Gee A, Dotti G, Bao R, Salem AH, Wang T, Brenner MK, Heslop HE, Wels WS, Hicks MJ, Gottschalk S, Ahmed N. Autologous HER2-specific CAR T cells after lymphodepletion for advanced sarcoma: a phase 1 trial. NATURE CANCER 2024; 5:880-894. [PMID: 38658775 PMCID: PMC11588040 DOI: 10.1038/s43018-024-00749-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/23/2024] [Indexed: 04/26/2024]
Abstract
In this prospective, interventional phase 1 study for individuals with advanced sarcoma, we infused autologous HER2-specific chimeric antigen receptor T cells (HER2 CAR T cells) after lymphodepletion with fludarabine (Flu) ± cyclophosphamide (Cy): 1 × 108 T cells per m2 after Flu (cohort A) or Flu/Cy (cohort B) and 1 × 108 CAR+ T cells per m2 after Flu/Cy (cohort C). The primary outcome was assessment of safety of one dose of HER2 CAR T cells after lymphodepletion. Determination of antitumor responses was the secondary outcome. Thirteen individuals were treated in 14 enrollments, and seven received multiple infusions. HER2 CAR T cells expanded after 19 of 21 infusions. Nine of 12 individuals in cohorts A and B developed grade 1-2 cytokine release syndrome. Two individuals in cohort C experienced dose-limiting toxicity with grade 3-4 cytokine release syndrome. Antitumor activity was observed with clinical benefit in 50% of individuals treated. The tumor samples analyzed showed spatial heterogeneity of immune cells and clustering by sarcoma type and by treatment response. Our results affirm HER2 as a CAR T cell target and demonstrate the safety of this therapeutic approach in sarcoma. ClinicalTrials.gov registration: NCT00902044 .
Collapse
Affiliation(s)
- Meenakshi Hegde
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA.
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA.
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
| | - Shoba Navai
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Christopher DeRenzo
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sujith K Joseph
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Khaled Sanber
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Mengfen Wu
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Ahmed Z Gad
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Katherine A Janeway
- Department of Pediatrics, Boston Children's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Matthew Campbell
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Dolores Mullikin
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Zeid Nawas
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Catherine Robertson
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Pretty R Mathew
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Huimin Zhang
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Birju Mehta
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Raksha R Bhat
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Angela Major
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Ankita Shree
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Claudia Gerken
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Mamta Kalra
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Rikhia Chakraborty
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Sachin G Thakkar
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Olga Dakhova
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Vita S Salsman
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Bambi Grilley
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Natalia Lapteva
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Adrian Gee
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Gianpietro Dotti
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Riyue Bao
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | | | - Tao Wang
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Malcolm K Brenner
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Helen E Heslop
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Winfried S Wels
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany
| | - M John Hicks
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Stephen Gottschalk
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Nabil Ahmed
- Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA.
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, TX, USA.
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
12
|
Deyell RJ, Shen Y, Titmuss E, Dixon K, Williamson LM, Pleasance E, Nelson JMT, Abbasi S, Krzywinski M, Armstrong L, Bonakdar M, Ch'ng C, Chuah E, Dunham C, Fok A, Jones M, Lee AF, Ma Y, Moore RA, Mungall AJ, Mungall KL, Rogers PC, Schrader KA, Virani A, Wee K, Young SS, Zhao Y, Jones SJM, Laskin J, Marra MA, Rassekh SR. Whole genome and transcriptome integrated analyses guide clinical care of pediatric poor prognosis cancers. Nat Commun 2024; 15:4165. [PMID: 38755180 PMCID: PMC11099106 DOI: 10.1038/s41467-024-48363-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 04/29/2024] [Indexed: 05/18/2024] Open
Abstract
The role for routine whole genome and transcriptome analysis (WGTA) for poor prognosis pediatric cancers remains undetermined. Here, we characterize somatic mutations, structural rearrangements, copy number variants, gene expression, immuno-profiles and germline cancer predisposition variants in children and adolescents with relapsed, refractory or poor prognosis malignancies who underwent somatic WGTA and matched germline sequencing. Seventy-nine participants with a median age at enrollment of 8.8 y (range 6 months to 21.2 y) are included. Germline pathogenic/likely pathogenic variants are identified in 12% of participants, of which 60% were not known prior. Therapeutically actionable variants are identified by targeted gene report and whole genome in 32% and 62% of participants, respectively, and increase to 96% after integrating transcriptome analyses. Thirty-two molecularly informed therapies are pursued in 28 participants with 54% achieving a clinical benefit rate; objective response or stable disease ≥6 months. Integrated WGTA identifies therapeutically actionable variants in almost all tumors and are directly translatable to clinical care of children with poor prognosis cancers.
Collapse
Affiliation(s)
- Rebecca J Deyell
- Department of Pediatrics, BC Children's Hospital and Research Institute, Vancouver, BC, Canada.
| | - Yaoqing Shen
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Emma Titmuss
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Katherine Dixon
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Laura M Williamson
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Erin Pleasance
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Jessica M T Nelson
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Sanna Abbasi
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Martin Krzywinski
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Linlea Armstrong
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Melika Bonakdar
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Carolyn Ch'ng
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Eric Chuah
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Chris Dunham
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Alexandra Fok
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Martin Jones
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Anna F Lee
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yussanne Ma
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Richard A Moore
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Andrew J Mungall
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Karen L Mungall
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Paul C Rogers
- Department of Pediatrics, BC Children's Hospital and Research Institute, Vancouver, BC, Canada
| | - Kasmintan A Schrader
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Alice Virani
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Kathleen Wee
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Sean S Young
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Cancer Genetics and Genomics Laboratory, Department of Pathology and Laboratory Medicine, BC Cancer, Vancouver, Canada
| | - Yongjun Zhao
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Janessa Laskin
- Department of Medical Oncology, BC Cancer, Vancouver, BC, Canada
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Shahrad R Rassekh
- Department of Pediatrics, BC Children's Hospital and Research Institute, Vancouver, BC, Canada.
| |
Collapse
|
13
|
Shaw TI, Wagner J, Tian L, Wickman E, Poudel S, Wang J, Paul R, Koo SC, Lu M, Sheppard H, Fan Y, O'Neill FH, Lau CC, Zhou X, Zhang J, Gottschalk S. Discovery of immunotherapy targets for pediatric solid and brain tumors by exon-level expression. Nat Commun 2024; 15:3732. [PMID: 38702309 PMCID: PMC11068777 DOI: 10.1038/s41467-024-47649-y] [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: 11/13/2023] [Accepted: 04/09/2024] [Indexed: 05/06/2024] Open
Abstract
Immunotherapy with chimeric antigen receptor T cells for pediatric solid and brain tumors is constrained by available targetable antigens. Cancer-specific exons present a promising reservoir of targets; however, these have not been explored and validated systematically in a pan-cancer fashion. To identify cancer specific exon targets, here we analyze 1532 RNA-seq datasets from 16 types of pediatric solid and brain tumors for comparison with normal tissues using a newly developed workflow. We find 2933 exons in 157 genes encoding proteins of the surfaceome or matrisome with high cancer specificity either at the gene (n = 148) or the alternatively spliced isoform (n = 9) level. Expression of selected alternatively spliced targets, including the EDB domain of fibronectin 1, and gene targets, such as COL11A1, are validated in pediatric patient derived xenograft tumors. We generate T cells expressing chimeric antigen receptors specific for the EDB domain or COL11A1 and demonstrate that these have antitumor activity. The full target list, explorable via an interactive web portal ( https://cseminer.stjude.org/ ), provides a rich resource for developing immunotherapy of pediatric solid and brain tumors using gene or AS targets with high expression specificity in cancer.
Collapse
Affiliation(s)
- Timothy I Shaw
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Jessica Wagner
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Liqing Tian
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Elizabeth Wickman
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Suresh Poudel
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jian Wang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Robin Paul
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Selene C Koo
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Meifen Lu
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Heather Sheppard
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yiping Fan
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Francis H O'Neill
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Ching C Lau
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
- Connecticut Children's Medical Center, Hartford, CT, 06106, USA
- University of Connecticut School of Medicine, Farmington, CT, 06032, USA
| | - Xin Zhou
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
14
|
Shao W, Yao Y, Yang L, Li X, Ge T, Zheng Y, Zhu Q, Ge S, Gu X, Jia R, Song X, Zhuang A. Novel insights into TCR-T cell therapy in solid neoplasms: optimizing adoptive immunotherapy. Exp Hematol Oncol 2024; 13:37. [PMID: 38570883 PMCID: PMC10988985 DOI: 10.1186/s40164-024-00504-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/21/2024] [Indexed: 04/05/2024] Open
Abstract
Adoptive immunotherapy in the T cell landscape exhibits efficacy in cancer treatment. Over the past few decades, genetically modified T cells, particularly chimeric antigen receptor T cells, have enabled remarkable strides in the treatment of hematological malignancies. Besides, extensive exploration of multiple antigens for the treatment of solid tumors has led to clinical interest in the potential of T cells expressing the engineered T cell receptor (TCR). TCR-T cells possess the capacity to recognize intracellular antigen families and maintain the intrinsic properties of TCRs in terms of affinity to target epitopes and signal transduction. Recent research has provided critical insight into their capability and therapeutic targets for multiple refractory solid tumors, but also exposes some challenges for durable efficacy. In this review, we describe the screening and identification of available tumor antigens, and the acquisition and optimization of TCRs for TCR-T cell therapy. Furthermore, we summarize the complete flow from laboratory to clinical applications of TCR-T cells. Last, we emerge future prospects for improving therapeutic efficacy in cancer world with combination therapies or TCR-T derived products. In conclusion, this review depicts our current understanding of TCR-T cell therapy in solid neoplasms, and provides new perspectives for expanding its clinical applications and improving therapeutic efficacy.
Collapse
Affiliation(s)
- Weihuan Shao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Yiran Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Ludi Yang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Xiaoran Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Tongxin Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Yue Zheng
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Qiuyi Zhu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Xiang Gu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China.
| | - Xin Song
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China.
| | - Ai Zhuang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
15
|
Belgiovine C, Mebelli K, Raffaele A, De Cicco M, Rotella J, Pedrazzoli P, Zecca M, Riccipetitoni G, Comoli P. Pediatric Solid Cancers: Dissecting the Tumor Microenvironment to Improve the Results of Clinical Immunotherapy. Int J Mol Sci 2024; 25:3225. [PMID: 38542199 PMCID: PMC10970338 DOI: 10.3390/ijms25063225] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/26/2024] [Accepted: 03/08/2024] [Indexed: 01/03/2025] Open
Abstract
Despite advances in their diagnosis and treatment, pediatric cancers remain among the leading causes of death in childhood. The development of immunotherapies and other forms of targeted therapies has significantly changed the prognosis of some previously incurable cancers in the adult population. However, so far, the results in pediatric cohorts are disappointing, which is mainly due to differences in tumor biology, including extreme heterogeneity and a generally low tumor mutational burden. A central role in the limited efficacy of immunotherapeutic approaches is played by the peculiar characteristics of the tumor microenvironment (TME) in pediatric cancer, with the scarcity of tumor infiltration by T cells and the abundance of stromal cells endowed with lymphocyte suppressor and tumor-growth-promoting activity. Thus, progress in the treatment of pediatric solid tumors will likely be influenced by the ability to modify the TME while delivering novel, more effective therapeutic agents. In this review, we will describe the TME composition in pediatric solid tumors and illustrate recent advances in treatment for the modulation of immune cells belonging to the TME.
Collapse
Affiliation(s)
- Cristina Belgiovine
- Dipartimento di Scienze Clinico-Chirurgiche, Diagnostiche e Pediatriche, University of Pavia, 27100 Pavia, Italy
- SC Chirurgia Pediatrica, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Kristiana Mebelli
- SC Chirurgia Pediatrica, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Alessandro Raffaele
- SC Chirurgia Pediatrica, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Marica De Cicco
- SSD Cell Factory e Center for Advanced Therapies, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Jessica Rotella
- SC Pediatric Hematology/Oncology, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Paolo Pedrazzoli
- Medical Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
- Department of Internal Medicine, University of Pavia, 27100 Pavia, Italy
| | - Marco Zecca
- SC Pediatric Hematology/Oncology, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Giovanna Riccipetitoni
- Dipartimento di Scienze Clinico-Chirurgiche, Diagnostiche e Pediatriche, University of Pavia, 27100 Pavia, Italy
- SC Chirurgia Pediatrica, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Patrizia Comoli
- SSD Cell Factory e Center for Advanced Therapies, Department of Woman and Child Health, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| |
Collapse
|
16
|
Schober SJ, Thiede M, Gassmann H, von Ofen AJ, Knoch P, Eck J, Prexler C, Kordass-Wally C, Hauer J, Burdach S, Holm PS, Thiel U. TCR-transgenic T cells and YB-1-based oncolytic virotherapy improve survival in a preclinical Ewing sarcoma xenograft mouse model. Front Immunol 2024; 15:1330868. [PMID: 38318175 PMCID: PMC10839048 DOI: 10.3389/fimmu.2024.1330868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/03/2024] [Indexed: 02/07/2024] Open
Abstract
Background Ewing sarcoma (EwS) is an aggressive and highly metastatic bone and soft tissue tumor in pediatric patients and young adults. Cure rates are low when patients present with metastatic or relapsed disease. Therefore, innovative therapy approaches are urgently needed. Cellular- and oncolytic virus-based immunotherapies are on the rise for solid cancers. Methods Here, we assess the combination of EwS tumor-associated antigen CHM1319-specific TCR-transgenic CD8+ T cells and the YB-1-driven (i.e. E1A13S-deleted) oncolytic adenovirus XVir-N-31 in vitro and in a xenograft mouse model for antitumor activity and immunostimulatory properties. Results In vitro both approaches specifically kill EwS cell lines in a synergistic manner over controls. This effect was confirmed in vivo, with increased survival using the combination therapy. Further in vitro analyses of immunogenic cell death and antigen presentation confirmed immunostimulatory properties of virus-infected EwS tumor cells. As dendritic cell maturation was also increased by XVir-N-31, we observed superior proliferation of CHM1319-specific TCR-transgenic CD8+ T cells only in virus-tested conditions, emphasizing the superior immune-activating potential of XVir-N-31. Conclusion Our data prove synergistic antitumor effects in vitro and superior tumor control in a preclinical xenograft setting. Combination strategies of EwS-redirected T cells and YB-1-driven virotherapy are a highly promising immunotherapeutic approach for EwS and warrant further evaluation in a clinical setting.
Collapse
Affiliation(s)
- Sebastian J. Schober
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Melanie Thiede
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Hendrik Gassmann
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Anna Josefine von Ofen
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Pia Knoch
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Jennifer Eck
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Carolin Prexler
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Corazon Kordass-Wally
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Julia Hauer
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Stefan Burdach
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Pathology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Per Sonne Holm
- Department of Urology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Department of Oral and Maxillofacial Surgery, Medical University Innsbruck, Innsbruck, Austria
| | - Uwe Thiel
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| |
Collapse
|
17
|
Wu C, Tan J, Shen H, Deng C, Kleber C, Osterhoff G, Schopow N. Exploring the relationship between metabolism and immune microenvironment in osteosarcoma based on metabolic pathways. J Biomed Sci 2024; 31:4. [PMID: 38212768 PMCID: PMC10785352 DOI: 10.1186/s12929-024-00999-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 01/02/2024] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND Metabolic remodeling and changes in tumor immune microenvironment (TIME) in osteosarcoma are important factors affecting prognosis and treatment. However, the relationship between metabolism and TIME needs to be further explored. METHODS RNA-Seq data and clinical information of 84 patients with osteosarcoma from the TARGET database and an independent cohort from the GEO database were included in this study. The activity of seven metabolic super-pathways and immune infiltration levels were inferred in osteosarcoma patients. Metabolism-related genes (MRGs) were identified and different metabolic clusters and MRG-related gene clusters were identified using unsupervised clustering. Then the TIME differences between the different clusters were compared. In addition, an MRGs-based risk model was constructed and the role of a key risk gene, ST3GAL4, in osteosarcoma cells was explored using molecular biological experiments. RESULTS This study revealed four key metabolic pathways in osteosarcoma, with vitamin and cofactor metabolism being the most relevant to prognosis and to TIME. Two metabolic pathway-related clusters (C1 and C2) were identified, with some differences in immune activating cell infiltration between the two clusters, and C2 was more likely to respond to two chemotherapeutic agents than C1. Three MRG-related gene clusters (GC1-3) were also identified, with significant differences in prognosis among the three clusters. GC2 and GC3 had higher immune cell infiltration than GC1. GC3 is most likely to respond to immune checkpoint blockade and to three commonly used clinical drugs. A metabolism-related risk model was developed and validated. The risk model has strong prognostic predictive power and the low-risk group has a higher level of immune infiltration than the high-risk group. Knockdown of ST3GAL4 significantly inhibited proliferation, migration, invasion and glycolysis of osteosarcoma cells and inhibited the M2 polarization of macrophages. CONCLUSION The metabolism of vitamins and cofactors is an important prognostic regulator of TIME in osteosarcoma, MRG-related gene clusters can well reflect changes in osteosarcoma TIME and predict chemotherapy and immunotherapy response. The metabolism-related risk model may serve as a useful prognostic predictor. ST3GAL4 plays a critical role in the progression, glycolysis, and TIME of osteosarcoma cells.
Collapse
Affiliation(s)
- Changwu Wu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jun Tan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Hong Shen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Chao Deng
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Christian Kleber
- Sarcoma Center, Department of Orthopedics, Trauma and Plastic Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Georg Osterhoff
- Sarcoma Center, Department of Orthopedics, Trauma and Plastic Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Nikolas Schopow
- Sarcoma Center, Department of Orthopedics, Trauma and Plastic Surgery, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
18
|
Shaw TI, Wagner J, Tian L, Wickman E, Poudel S, Wang J, Paul R, Koo SC, Lu M, Sheppard H, Fan Y, O’Neil F, Lau CC, Zhou X, Zhang J, Gottschalk S. Discovery of immunotherapy targets for pediatric solid and brain tumors by exon-level expression. RESEARCH SQUARE 2024:rs.3.rs-3821632. [PMID: 38260279 PMCID: PMC10802740 DOI: 10.21203/rs.3.rs-3821632/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: 01/24/2024]
Abstract
Immunotherapy with CAR T cells for pediatric solid and brain tumors is constrained by available targetable antigens. Cancer-specific exons (CSE) present a promising reservoir of targets; however, these have not been explored and validated systematically in a pan-cancer fashion. To identify CSE targets, we analyzed 1,532 RNA-seq datasets from 16 types of pediatric solid and brain tumors for comparison with normal tissues using a newly developed workflow. We found 2,933 exons in 157 genes encoding proteins of the surfaceome or matrisome with high cancer specificity either at the gene (n=148) or the alternatively spliced (AS) isoform (n=9) level. Expression of selected AS targets, including the EDB domain of FN1 (EDB), and gene targets, such as COL11A1, were validated in pediatric PDX tumors. We generated CAR T cells specific to EDB or COL11A1 and demonstrated that COL11A1-CAR T-cells have potent antitumor activity. The full target list, explorable via an interactive web portal (https://cseminer.stjude.org/), provides a rich resource for developing immunotherapy of pediatric solid and brain tumors using gene or AS targets with high expression specificity in cancer.
Collapse
Affiliation(s)
- Timothy I Shaw
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Jessica Wagner
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Liqing Tian
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Elizabeth Wickman
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Suresh Poudel
- Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Jian Wang
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Robin Paul
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Selene C. Koo
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Meifen Lu
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Heather Sheppard
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Yiping Fan
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Francis O’Neil
- The Jackson Laboratory Cancer Center, Farmington, CT, USA
| | - Ching C. Lau
- The Jackson Laboratory Cancer Center, Farmington, CT, USA
| | - Xin Zhou
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
19
|
Valtingojer I, Lièvre S, Bordes P, Paranjpe K, Thompson W, Shah S, Fantin V, Jacquemet-Ross W, Adamson PC. Collaborative Innovations in Childhood Cancer Therapies. Handb Exp Pharmacol 2024; 286:33-50. [PMID: 39177748 DOI: 10.1007/164_2024_725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
The outcome for children with cancer has improved significantly over the past 60 years, with more than 80% of patients today becoming 5-year survivors. Despite this progress, cancer remains the leading cause of death from disease in children in the United States and Europe, with significant short- and long-term toxicity of treatment continuing to impact most children. While the past 15 years have witnessed dramatic scientific innovation for certain cancers in adult patients, pediatric cancer treatment innovation lags increasingly behind. To help bridge the adult-pediatric therapeutic development gap, collaborative efforts are essential among stakeholders within and outside the pediatric oncology community. Prioritizing collaboration in areas such as cancer characterization, target identification and validation, drug discovery, and approaches to currently "undruggable" targets is imperative to improving the outcomes for children with cancer.
Collapse
Affiliation(s)
| | - Sasha Lièvre
- Department of Oncology, R&D, Sanofi, Vitry-sur-Seine, France
| | - Philippe Bordes
- Department of Alliance Management and Public Private Partnerships, Sanofi, Gentilly, France
| | - Krupa Paranjpe
- Department of Oncology Medical Affairs, Sanofi, Cambridge, MA, USA
| | | | - Sachin Shah
- Department of Oncology Medical Affairs, Sanofi, Cambridge, MA, USA
| | | | | | | |
Collapse
|
20
|
Tetens AR, Martin AM, Arnold A, Novak OV, Idrizi A, Tryggvadottir R, Craig-Schwartz J, Liapodimitri A, Lunsford K, Barbato MI, Eberhart CG, Resnick AC, Raabe EH, Koldobskiy MA. DNA methylation landscapes in DIPG reveal methylome variability that can be modified pharmacologically. Neurooncol Adv 2024; 6:vdae023. [PMID: 38468866 PMCID: PMC10926944 DOI: 10.1093/noajnl/vdae023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024] Open
Abstract
Background Diffuse intrinsic pontine glioma (DIPG) is a uniformly lethal brainstem tumor of childhood, driven by histone H3 K27M mutation and resultant epigenetic dysregulation. Epigenomic analyses of DIPG have shown global loss of repressive chromatin marks accompanied by DNA hypomethylation. However, studies providing a static view of the epigenome do not adequately capture the regulatory underpinnings of DIPG cellular heterogeneity and plasticity. Methods To address this, we performed whole-genome bisulfite sequencing on a large panel of primary DIPG specimens and applied a novel framework for analysis of DNA methylation variability, permitting the derivation of comprehensive genome-wide DNA methylation potential energy landscapes that capture intrinsic epigenetic variation. Results We show that DIPG has a markedly disordered epigenome with increasingly stochastic DNA methylation at genes regulating pluripotency and developmental identity, potentially enabling cells to sample diverse transcriptional programs and differentiation states. The DIPG epigenetic landscape was responsive to treatment with the hypomethylating agent decitabine, which produced genome-wide demethylation and reduced the stochasticity of DNA methylation at active enhancers and bivalent promoters. Decitabine treatment elicited changes in gene expression, including upregulation of immune signaling such as the interferon response, STING, and MHC class I expression, and sensitized cells to the effects of histone deacetylase inhibition. Conclusions This study provides a resource for understanding the epigenetic instability that underlies DIPG heterogeneity. It suggests the application of epigenetic therapies to constrain the range of epigenetic states available to DIPG cells, as well as the use of decitabine in priming for immune-based therapies.
Collapse
Affiliation(s)
- Ashley R Tetens
- Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Allison M Martin
- Pediatric Hematology-Oncology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Antje Arnold
- Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Orlandi V Novak
- Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Adrian Idrizi
- Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rakel Tryggvadottir
- Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jordyn Craig-Schwartz
- Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Athanasia Liapodimitri
- Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kayleigh Lunsford
- Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael I Barbato
- Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Charles G Eberhart
- Neuropathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Adam C Resnick
- Center for Data-Driven Discovery in Biomedicine, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Division of Neurosurgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Eric H Raabe
- Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Neuropathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael A Koldobskiy
- Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
21
|
Figueiredo D, Cruz RGB, Normando AGC, Granato DC, Busso-Lopes AF, Carnielli CM, De Rossi T, Paes Leme AF. Peptidomics Strategies to Evaluate Cancer Diagnosis, Prognosis, and Treatment. Methods Mol Biol 2024; 2758:401-423. [PMID: 38549027 DOI: 10.1007/978-1-0716-3646-6_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Peptides have potential bioactive functions, and the peptidomics landscape has been broadly investigated for various diseases, including cancer. In this chapter, we reviewed the past four years of literature available and selected 16 peer-reviewed publications exploring peptidomics in diagnosis, prognosis, and treatment in cancer research. We highlighted their main aims, mass spectrometry-based peptidomics, multi-omics, data-driven and in silico strategies, functional assays, and clinical applications. Moreover, we underscored several levels of difficulties in translating the peptidomics findings to clinical practice, aiming to learn with the accumulated knowledge and guide upcoming studies. Finally, this review reinforces the peptidomics robustness in discovering potential candidates for monitoring the several stages of cancer disease and therapeutic treatment, leveraging the management of cancer patients in the future.
Collapse
Affiliation(s)
- Daniella Figueiredo
- Laboratório Nacional de Biociências, LNBio, Centro Nacional de Pesquisa em Energia e Materiais, CNPEM, Campinas, SP, Brazil
| | - Rodrigo G B Cruz
- Laboratório Nacional de Biociências, LNBio, Centro Nacional de Pesquisa em Energia e Materiais, CNPEM, Campinas, SP, Brazil
| | - Ana Gabriela Costa Normando
- Laboratório Nacional de Biociências, LNBio, Centro Nacional de Pesquisa em Energia e Materiais, CNPEM, Campinas, SP, Brazil
| | - Daniela C Granato
- Laboratório Nacional de Biociências, LNBio, Centro Nacional de Pesquisa em Energia e Materiais, CNPEM, Campinas, SP, Brazil
| | - Ariane F Busso-Lopes
- Laboratório Nacional de Biociências, LNBio, Centro Nacional de Pesquisa em Energia e Materiais, CNPEM, Campinas, SP, Brazil
| | - Carolina M Carnielli
- Laboratório Nacional de Biociências, LNBio, Centro Nacional de Pesquisa em Energia e Materiais, CNPEM, Campinas, SP, Brazil
| | - Tatiane De Rossi
- Laboratório Nacional de Biociências, LNBio, Centro Nacional de Pesquisa em Energia e Materiais, CNPEM, Campinas, SP, Brazil
| | - Adriana Franco Paes Leme
- Laboratório Nacional de Biociências, LNBio, Centro Nacional de Pesquisa em Energia e Materiais, CNPEM, Campinas, SP, Brazil.
| |
Collapse
|
22
|
Ebegboni VJ, Jones TL, Brownmiller T, Zhao PX, Pehrsson EC, Rajan SS, Caplen NJ. ETS1, a target gene of the EWSR1::FLI1 fusion oncoprotein, regulates the expression of the focal adhesion protein TENSIN3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.572864. [PMID: 38187702 PMCID: PMC10769395 DOI: 10.1101/2023.12.21.572864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The mechanistic basis for the metastasis of Ewing sarcomas remains poorly understood, as these tumors harbor few mutations beyond the chromosomal translocation that initiates the disease. Instead, the epigenome of Ewing sarcoma (EWS) cells reflects the regulatory state of genes associated with the DNA binding activity of the fusion oncoproteins EWSR1::FLI1 or EWSR1::ERG. In this study, we examined the EWSR1::FLI1/ERG's repression of transcription factor genes, concentrating on those that exhibit a broader range of expression in tumors than in EWS cell lines. Focusing on one of these target genes, ETS1, we detected EWSR1::FLI1 binding and an H3K27me3 repressive mark at this locus. Depletion of EWSR1::FLI1 results in ETS1's binding of promoter regions, substantially altering the transcriptome of EWS cells, including the upregulation of the gene encoding TENSIN3 (TNS3), a focal adhesion protein. EWS cell lines expressing ETS1 (CRISPRa) exhibited increased TNS3 expression and enhanced movement compared to control cells. The cytoskeleton of control cells and ETS1-activated EWS cell lines also differed. Specifically, control cells exhibited a distributed vinculin signal and a network-like organization of F-actin. In contrast, ETS1-activated EWS cells showed an accumulation of vinculin and F-actin towards the plasma membrane. Interestingly, the phenotype of ETS1-activated EWS cell lines depleted of TNS3 resembled the phenotype of the control cells. Critically, these findings have clinical relevance as TNS3 expression in EWS tumors positively correlates with that of ETS1.
Collapse
Affiliation(s)
- Vernon Justice Ebegboni
- Functional Genetics Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tamara L Jones
- Functional Genetics Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tayvia Brownmiller
- Functional Genetics Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Patrick X Zhao
- Omics Bioinformatics Facility, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Erica C Pehrsson
- Omics Bioinformatics Facility, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Soumya Sundara Rajan
- Functional Genetics Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Natasha J Caplen
- Functional Genetics Section, Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
23
|
Fujii S, Ishida M, Komura K, Nishimura K, Tsujino T, Saito T, Taniguchi Y, Murakawa T, Azuma H, Hirose Y. Expression of Preferentially Expressed Antigen in Melanoma, a Cancer/Testis Antigen, in Carcinoma In Situ of the Urinary Tract. Diagnostics (Basel) 2023; 13:3636. [PMID: 38132219 PMCID: PMC10742698 DOI: 10.3390/diagnostics13243636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/07/2023] [Accepted: 12/09/2023] [Indexed: 12/23/2023] Open
Abstract
Carcinoma in situ (CIS) of the urinary tract comprises 1-3% of all urothelial malignancies and is often a precursor to muscle-invasive urothelial carcinoma (UC). This study aimed to examine the expression profiles of preferentially expressed antigen in melanoma (PRAME), a cancer/testis antigen, and assess its diagnostic and therapeutic applications in CIS, given that its expression in UC has been minimally studied and has not yet been analyzed in CIS. We selected consecutive patients with CIS who underwent biopsy and/or transurethral tumor resection at the Osaka Medical and Pharmaceutical University Hospital. Immunohistochemical staining for PRAME and p53 was performed. Overall, 53 patients with CIS (6 females and 47 males) were included. Notably, PRAME expression was observed in 23 of the 53 patients (43.4%), whereas it was absent in the non-neoplastic urothelial epithelium. Furthermore, no correlation was found between PRAME expression and aberrant p53 expression. Therefore, PRAME expression may serve as a useful marker for CIS of the urinary tract. Furthermore, PRAME may be a candidate for the novel therapeutic target for standard treatment-refractory CIS patients.
Collapse
Affiliation(s)
- Shota Fujii
- Department of Pathology, Osaka Medical and Pharmaceutical University, 2-7, Daigaku-machi, Takatsuki City 569-8686, Osaka, Japan; (S.F.)
| | - Mitsuaki Ishida
- Department of Pathology, Osaka Medical and Pharmaceutical University, 2-7, Daigaku-machi, Takatsuki City 569-8686, Osaka, Japan; (S.F.)
| | - Kazumasa Komura
- Department of Urology, Osaka Medical and Pharmaceutical University, 2-7, Daigaku-machi, Takatsuki City 569-8686, Osaka, Japan
- Translational Research Program, Osaka Medical and Pharmaceutical University, 2-7, Daigaku-machi, Takatsuki City 569-8686, Osaka, Japan
| | - Kazuki Nishimura
- Department of Urology, Osaka Medical and Pharmaceutical University, 2-7, Daigaku-machi, Takatsuki City 569-8686, Osaka, Japan
| | - Takuya Tsujino
- Department of Urology, Osaka Medical and Pharmaceutical University, 2-7, Daigaku-machi, Takatsuki City 569-8686, Osaka, Japan
| | - Tomohito Saito
- Department of Thoracic Surgery, Kansai Medical University, 2-5-1, Shinmachi, Hirakata 573-1010, Osaka, Japan
| | - Yohei Taniguchi
- Department of Thoracic Surgery, Kansai Medical University, 2-5-1, Shinmachi, Hirakata 573-1010, Osaka, Japan
| | - Tomohiro Murakawa
- Department of Thoracic Surgery, Kansai Medical University, 2-5-1, Shinmachi, Hirakata 573-1010, Osaka, Japan
| | - Haruhito Azuma
- Department of Urology, Osaka Medical and Pharmaceutical University, 2-7, Daigaku-machi, Takatsuki City 569-8686, Osaka, Japan
| | - Yoshinobu Hirose
- Department of Pathology, Osaka Medical and Pharmaceutical University, 2-7, Daigaku-machi, Takatsuki City 569-8686, Osaka, Japan; (S.F.)
| |
Collapse
|
24
|
McKay-Corkum GB, Collins VJ, Yeung C, Ito T, Issaq SH, Holland D, Vulikh K, Zhang Y, Lee U, Lei H, Mendoza A, Shern JF, Yohe ME, Yamamoto K, Wilson K, Ji J, Karim BO, Thomas CJ, Krishna MC, Neckers LM, Heske CM. Inhibition of NAD+-Dependent Metabolic Processes Induces Cellular Necrosis and Tumor Regression in Rhabdomyosarcoma Models. Clin Cancer Res 2023; 29:4479-4491. [PMID: 37616468 PMCID: PMC10841338 DOI: 10.1158/1078-0432.ccr-23-0200] [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: 02/24/2023] [Revised: 06/23/2023] [Accepted: 08/22/2023] [Indexed: 08/26/2023]
Abstract
PURPOSE Deregulated metabolism in cancer cells represents a vulnerability that may be therapeutically exploited to benefit patients. One such target is nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in the NAD+ salvage pathway. NAMPT is necessary for efficient NAD+ production and may be exploited in cells with increased metabolic demands. We have identified NAMPT as a dependency in rhabdomyosarcoma (RMS), a malignancy for which novel therapies are critically needed. Here we describe the effect of NAMPT inhibition on RMS proliferation and metabolism in vitro and in vivo. EXPERIMENTAL DESIGN Assays of proliferation and cell death were used to determine the effects of pharmacologic NAMPT inhibition in a panel of ten molecularly diverse RMS cell lines. Mechanism of the clinical NAMPTi OT-82 was determined using measures of NAD+ and downstream NAD+-dependent functions, including energy metabolism. We used orthotopic xenograft models to examine tolerability, efficacy, and drug mechanism in vivo. RESULTS Across all ten RMS cell lines, OT-82 depleted NAD+ and inhibited cell growth at concentrations ≤1 nmol/L. Significant impairment of glycolysis was a universal finding, with some cell lines also exhibiting diminished oxidative phosphorylation. Most cell lines experienced profound depletion of ATP with subsequent irreversible necrotic cell death. Importantly, loss of NAD and glycolytic activity were confirmed in orthotopic in vivo models, which exhibited complete tumor regressions with OT-82 treatment delivered on the clinical schedule. CONCLUSIONS RMS is highly vulnerable to NAMPT inhibition. These findings underscore the need for further clinical study of this class of agents for this malignancy.
Collapse
Affiliation(s)
- Grace B. McKay-Corkum
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)
| | - Victor J. Collins
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)
| | - Choh Yeung
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)
| | - Takeshi Ito
- Urologic Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)
| | - Sameer H. Issaq
- Urologic Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)
| | - David Holland
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health (NIH)
| | - Ksenia Vulikh
- Molecular Histopathology Lab, Frederick National Laboratory for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH)
| | - Yiping Zhang
- National Clinical Target Validation Laboratory, Frederick National Laboratory for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH)
| | - Unsun Lee
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)
| | - Haiyan Lei
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)
| | - Arnulfo Mendoza
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)
| | - Jack F. Shern
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)
| | - Marielle E. Yohe
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)
| | - Kazutoshi Yamamoto
- Radiation Biology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)
| | - Kelli Wilson
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health (NIH)
| | - Jiuping Ji
- National Clinical Target Validation Laboratory, Frederick National Laboratory for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH)
| | - Baktiar O. Karim
- Molecular Histopathology Lab, Frederick National Laboratory for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH)
| | - Craig J. Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health (NIH)
| | - Murali C. Krishna
- Radiation Biology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)
| | - Leonard M. Neckers
- Urologic Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)
| | - Christine M. Heske
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH)
| |
Collapse
|
25
|
Tian M, Wei JS, Shivaprasad N, Highfill SL, Gryder BE, Milewski D, Brown GT, Moses L, Song H, Wu JT, Azorsa P, Kumar J, Schneider D, Chou HC, Song YK, Rahmy A, Masih KE, Kim YY, Belyea B, Linardic CM, Dropulic B, Sullivan PM, Sorensen PH, Dimitrov DS, Maris JM, Mackall CL, Orentas RJ, Cheuk AT, Khan J. Preclinical development of a chimeric antigen receptor T cell therapy targeting FGFR4 in rhabdomyosarcoma. Cell Rep Med 2023; 4:101212. [PMID: 37774704 PMCID: PMC10591056 DOI: 10.1016/j.xcrm.2023.101212] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 06/12/2023] [Accepted: 09/06/2023] [Indexed: 10/01/2023]
Abstract
Pediatric patients with relapsed or refractory rhabdomyosarcoma (RMS) have dismal cure rates, and effective therapy is urgently needed. The oncogenic receptor tyrosine kinase fibroblast growth factor receptor 4 (FGFR4) is highly expressed in RMS and lowly expressed in healthy tissues. Here, we describe a second-generation FGFR4-targeting chimeric antigen receptor (CAR), based on an anti-human FGFR4-specific murine monoclonal antibody 3A11, as an adoptive T cell treatment for RMS. The 3A11 CAR T cells induced robust cytokine production and cytotoxicity against RMS cell lines in vitro. In contrast, a panel of healthy human primary cells failed to activate 3A11 CAR T cells, confirming the selectivity of 3A11 CAR T cells against tumors with high FGFR4 expression. Finally, we demonstrate that 3A11 CAR T cells are persistent in vivo and can effectively eliminate RMS tumors in two metastatic and two orthotopic models. Therefore, our study credentials CAR T cell therapy targeting FGFR4 to treat patients with RMS.
Collapse
Affiliation(s)
- Meijie Tian
- Genetics Branch, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Jun S Wei
- Genetics Branch, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Nityashree Shivaprasad
- Genetics Branch, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Steven L Highfill
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Berkley E Gryder
- Genetics Branch, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - David Milewski
- Genetics Branch, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - G Tom Brown
- Artificial Intelligence Resource, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Larry Moses
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Hannah Song
- Center for Cellular Engineering, Department of Transfusion Medicine, National Institutes of Health Clinical Center, Bethesda, MD 20892, USA
| | - Jerry T Wu
- Genetics Branch, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Peter Azorsa
- Genetics Branch, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Jeetendra Kumar
- Genetics Branch, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Dina Schneider
- Lentigen Corporation, Miltenyi Bioindustry, 1201 Clopper Road, Gaithersburg, MD 20878, USA
| | - Hsien-Chao Chou
- Genetics Branch, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Young K Song
- Genetics Branch, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Abdelrahman Rahmy
- Genetics Branch, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Katherine E Masih
- Genetics Branch, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA; Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Yong Yean Kim
- Genetics Branch, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Brian Belyea
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Corinne M Linardic
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Boro Dropulic
- Caring Cross, 708 Quince Orchard Road, Gaithersburg, MD 20878, USA
| | - Peter M Sullivan
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, 1100 Olive Way, Seattle, WA 98101, USA
| | - Poul H Sorensen
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
| | - Dimiter S Dimitrov
- University of Pittsburgh Department of Medicine, Pittsburgh, PA 15261, USA
| | - John M Maris
- Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Crystal L Mackall
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rimas J Orentas
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, 1100 Olive Way, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98101, USA
| | - Adam T Cheuk
- Genetics Branch, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA.
| | - Javed Khan
- Genetics Branch, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
26
|
Chen AP, Sharon E, O'Sullivan-Coyne G, Moore N, Foster JC, Hu JS, Van Tine BA, Conley AP, Read WL, Riedel RF, Burgess MA, Glod J, Davis EJ, Merriam P, Naqash AR, Fino KK, Miller BL, Wilsker DF, Begum A, Ferry-Galow KV, Deshpande HA, Schwartz GK, Ladle BH, Okuno SH, Beck JC, Chen JL, Takebe N, Fogli LK, Rosenberger CL, Parchment RE, Doroshow JH. Atezolizumab for Advanced Alveolar Soft Part Sarcoma. N Engl J Med 2023; 389:911-921. [PMID: 37672694 PMCID: PMC10729808 DOI: 10.1056/nejmoa2303383] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
BACKGROUND Alveolar soft part sarcoma (ASPS) is a rare soft-tissue sarcoma with a poor prognosis and no established therapy. Recently, encouraging responses to immune checkpoint inhibitors have been reported. METHODS We conducted an investigator-initiated, multicenter, single-group, phase 2 study of the anti-programmed death ligand 1 (PD-L1) agent atezolizumab in adult and pediatric patients with advanced ASPS. Atezolizumab was administered intravenously at a dose of 1200 mg (in patients ≥18 years of age) or 15 mg per kilogram of body weight with a 1200-mg cap (in patients <18 years of age) once every 21 days. Study end points included objective response, duration of response, and progression-free survival according to Response Evaluation Criteria in Solid Tumors (RECIST), version 1.1, as well as pharmacodynamic biomarkers of multistep drug action. RESULTS A total of 52 patients were evaluated. An objective response was observed in 19 of 52 patients (37%), with 1 complete response and 18 partial responses. The median time to response was 3.6 months (range, 2.1 to 19.1), the median duration of response was 24.7 months (range, 4.1 to 55.8), and the median progression-free survival was 20.8 months. Seven patients took a treatment break after 2 years of treatment, and their responses were maintained through the data-cutoff date. No treatment-related grade 4 or 5 adverse events were recorded. Responses were noted despite variable baseline expression of programmed death 1 and PD-L1. CONCLUSIONS Atezolizumab was effective at inducing sustained responses in approximately one third of patients with advanced ASPS. (Funded by the National Cancer Institute and others; ClinicalTrials.gov number, NCT03141684.).
Collapse
Affiliation(s)
- Alice P Chen
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Elad Sharon
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Geraldine O'Sullivan-Coyne
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Nancy Moore
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Jared C Foster
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - James S Hu
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Brian A Van Tine
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Anthony P Conley
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - William L Read
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Richard F Riedel
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Melissa A Burgess
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - John Glod
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Elizabeth J Davis
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Priscilla Merriam
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Abdul R Naqash
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Kristin K Fino
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Brandon L Miller
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Deborah F Wilsker
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Asma Begum
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Katherine V Ferry-Galow
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Hari A Deshpande
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Gary K Schwartz
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Brian H Ladle
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Scott H Okuno
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Jill C Beck
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - James L Chen
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Naoko Takebe
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Laura K Fogli
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Christina L Rosenberger
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - Ralph E Parchment
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| | - James H Doroshow
- From the Division of Cancer Treatment and Diagnosis (A.P. Chen, E.S., G.O.-C., N.M., J.C.F., A.R.N., N.T., L.K.F., C.L.R., J.H.D.) and the Center for Cancer Research (J.G., J.H.D.), National Cancer Institute, Bethesda, the Clinical Pharmacodynamics Biomarker Program, Applied and Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick (K.K.F., B.L.M., D.F.W., A.B., K.V.F.-G., R.E.P.), and the Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore (B.H.L.) - all in Maryland; the Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles (J.S.H.); the Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis (B.A.V.T.); the University of Texas M.D. Anderson Cancer Center, Houston (A.P. Conley); Emory University, Atlanta (W.L.R.); Duke Cancer Institute, Duke University Medical Center, Durham, NC (R.F.R.); University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh (M.A.B.); the Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville (E.J.D.); the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston (P.M.); the Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (H.A.D.); the Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York (G.K.S.); Mayo Clinic, Rochester, MN (S.H.O.); the Division of Pediatric Hematology-Oncology, University of Nebraska Medical Center, Omaha (J.C.B.); the Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus (J.L.C.); and Stephenson Cancer Center at the University of Oklahoma, Oklahoma City (A.R.N.)
| |
Collapse
|
27
|
Reed DR, Grohar P, Rubin E, Binitie O, Krailo M, Davis J, DuBois SG, Janeway KA. Children's Oncology Group's 2023 blueprint for research: Bone tumors. Pediatr Blood Cancer 2023; 70 Suppl 6:e30583. [PMID: 37501549 PMCID: PMC10499366 DOI: 10.1002/pbc.30583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 07/08/2023] [Indexed: 07/29/2023]
Abstract
The Children's Oncology Group (COG) Bone Tumor Committee is responsible for clinical trials and biological research on localized, metastatic, and recurrent osteosarcoma and Ewing sarcoma (EWS). Results of clinical trials in localized disease completed and published in the past 10 years have led to international standard-of-care chemotherapy for osteosarcoma and EWS. A recent focus on identifying disease subgroups has led to the identification of biological features associated with poor outcomes including the presence of circulating tumor DNA (ctDNA) at diagnosis, and specific genomic alterations-MYC amplification for osteosarcoma and STAG2 and TP53 mutation for EWS. Studies validating these potential biomarkers are under way. Clinical trials evaluating the addition of multitargeted kinase inhibitors, which are active in relapsed bone sarcomas, to standard chemotherapy are under way in osteosarcoma and planned in EWS. In addition, the Committee has data analyses and a clinical trial under way to evaluate approaches to local management of the primary tumor and metastatic sites. Given the rarity of bone sarcomas, we have prioritized international interactions and are in the process of forming an international data-sharing consortium to facilitate refinement of risk stratification and study of rare disease subtypes.
Collapse
Affiliation(s)
- Damon R Reed
- Department of Individualized Cancer Management, Moffitt Cancer Center, Tampa, Florida, USA
| | - Patrick Grohar
- Division of Oncology, Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elyssa Rubin
- Department of Oncology, Children's Hospital of Orange County, Orange, California, USA
| | - Odion Binitie
- Department of Sarcoma, Moffitt Cancer Center, Tampa, Florida, USA
| | - Mark Krailo
- Keck School of Medicine, University of Southern California and Children's Oncology Group, Monrovia, California, USA
| | - Jessica Davis
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Steven G DuBois
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts, USA
| | - Katherine A Janeway
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts, USA
| |
Collapse
|
28
|
Mannheimer JD, Tawa G, Gerhold D, Braisted J, Sayers CM, McEachron TA, Meltzer P, Mazcko C, Beck JA, LeBlanc AK. Transcriptional profiling of canine osteosarcoma identifies prognostic gene expression signatures with translational value for humans. Commun Biol 2023; 6:856. [PMID: 37591946 PMCID: PMC10435536 DOI: 10.1038/s42003-023-05208-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 08/03/2023] [Indexed: 08/19/2023] Open
Abstract
Canine osteosarcoma is increasingly recognized as an informative model for human osteosarcoma. Here we show in one of the largest clinically annotated canine osteosarcoma transcriptional datasets that two previously reported, as well as de novo gene signatures devised through single sample Gene Set Enrichment Analysis (ssGSEA), have prognostic utility in both human and canine patients. Shared molecular pathway alterations are seen in immune cell signaling and activation including TH1 and TH2 signaling, interferon signaling, and inflammatory responses. Virtual cell sorting to estimate immune cell populations within canine and human tumors showed similar trends, predominantly for macrophages and CD8+ T cells. Immunohistochemical staining verified the increased presence of immune cells in tumors exhibiting immune gene enrichment. Collectively these findings further validate naturally occurring osteosarcoma of the pet dog as a translationally relevant patient model for humans and improve our understanding of the immunologic and genomic landscape of the disease in both species.
Collapse
Affiliation(s)
- Joshua D Mannheimer
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Gregory Tawa
- Division of Preclinical Innovation, Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - David Gerhold
- Division of Preclinical Innovation, Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - John Braisted
- Division of Preclinical Innovation, Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Carly M Sayers
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Troy A McEachron
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Paul Meltzer
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christina Mazcko
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jessica A Beck
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amy K LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
29
|
Wu JT, Cheuk A, Isanogle K, Robinson C, Zhang X, Ceribelli M, Beck E, Shinn P, Klumpp-Thomas C, Wilson KM, McKnight C, Itkin Z, Sotome H, Hirai H, Calleja E, Wacheck V, Gouker B, Peer CJ, Corvalan N, Milewski D, Kim YY, Figg WD, Edmondson EF, Thomas CJ, Difilippantonio S, Wei JS, Khan J. Preclinical Evaluation of the FGFR-Family Inhibitor Futibatinib for Pediatric Rhabdomyosarcoma. Cancers (Basel) 2023; 15:4034. [PMID: 37627061 PMCID: PMC10452847 DOI: 10.3390/cancers15164034] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/25/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is the most common pediatric soft tissue sarcoma. Despite decades of clinical trials, the overall survival rate for patients with relapsed and metastatic disease remains below 30%, underscoring the need for novel treatments. FGFR4, a receptor tyrosine kinase that is overexpressed in RMS and mutationally activated in 10% of cases, is a promising target for treatment. Here, we show that futibatinib, an irreversible pan-FGFR inhibitor, inhibits the growth of RMS cell lines in vitro by inhibiting phosphorylation of FGFR4 and its downstream targets. Moreover, we provide evidence that the combination of futibatinib with currently used chemotherapies such as irinotecan and vincristine has a synergistic effect against RMS in vitro. However, in RMS xenograft models, futibatinib monotherapy and combination treatment have limited efficacy in delaying tumor growth and prolonging survival. Moreover, limited efficacy is only observed in a PAX3-FOXO1 fusion-negative (FN) RMS cell line with mutationally activated FGFR4, whereas little or no efficacy is observed in PAX3-FOXO1 fusion-positive (FP) RMS cell lines with FGFR4 overexpression. Alternative treatment modalities such as combining futibatinib with other kinase inhibitors or targeting FGFR4 with CAR T cells or antibody-drug conjugate may be more effective than the approaches tested in this study.
Collapse
Affiliation(s)
- Jerry T. Wu
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (J.T.W.); (A.C.); (D.M.); (Y.Y.K.); (J.S.W.)
| | - Adam Cheuk
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (J.T.W.); (A.C.); (D.M.); (Y.Y.K.); (J.S.W.)
| | - Kristine Isanogle
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (K.I.); (C.R.); (S.D.)
| | - Christina Robinson
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (K.I.); (C.R.); (S.D.)
| | - Xiaohu Zhang
- National Center for Advancing Translational Sciences, Rockville, MD 20850, USA; (X.Z.); (M.C.); (E.B.); (P.S.); (C.K.-T.); (K.M.W.); (C.M.); (Z.I.); (C.J.T.)
| | - Michele Ceribelli
- National Center for Advancing Translational Sciences, Rockville, MD 20850, USA; (X.Z.); (M.C.); (E.B.); (P.S.); (C.K.-T.); (K.M.W.); (C.M.); (Z.I.); (C.J.T.)
| | - Erin Beck
- National Center for Advancing Translational Sciences, Rockville, MD 20850, USA; (X.Z.); (M.C.); (E.B.); (P.S.); (C.K.-T.); (K.M.W.); (C.M.); (Z.I.); (C.J.T.)
| | - Paul Shinn
- National Center for Advancing Translational Sciences, Rockville, MD 20850, USA; (X.Z.); (M.C.); (E.B.); (P.S.); (C.K.-T.); (K.M.W.); (C.M.); (Z.I.); (C.J.T.)
| | - Carleen Klumpp-Thomas
- National Center for Advancing Translational Sciences, Rockville, MD 20850, USA; (X.Z.); (M.C.); (E.B.); (P.S.); (C.K.-T.); (K.M.W.); (C.M.); (Z.I.); (C.J.T.)
| | - Kelli M. Wilson
- National Center for Advancing Translational Sciences, Rockville, MD 20850, USA; (X.Z.); (M.C.); (E.B.); (P.S.); (C.K.-T.); (K.M.W.); (C.M.); (Z.I.); (C.J.T.)
| | - Crystal McKnight
- National Center for Advancing Translational Sciences, Rockville, MD 20850, USA; (X.Z.); (M.C.); (E.B.); (P.S.); (C.K.-T.); (K.M.W.); (C.M.); (Z.I.); (C.J.T.)
| | - Zina Itkin
- National Center for Advancing Translational Sciences, Rockville, MD 20850, USA; (X.Z.); (M.C.); (E.B.); (P.S.); (C.K.-T.); (K.M.W.); (C.M.); (Z.I.); (C.J.T.)
| | - Hiroshi Sotome
- Taiho Pharmaceutical Co., Ltd., Tsukuba 300-0034, Japan; (H.S.); (H.H.)
| | - Hiroshi Hirai
- Taiho Pharmaceutical Co., Ltd., Tsukuba 300-0034, Japan; (H.S.); (H.H.)
| | | | - Volker Wacheck
- Taiho Oncology, Princeton, NJ 08540, USA; (E.C.); (V.W.)
| | - Brad Gouker
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (K.I.); (C.R.); (S.D.)
| | - Cody J. Peer
- Clinical Pharmacology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA (N.C.)
| | - Natalia Corvalan
- Clinical Pharmacology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA (N.C.)
| | - David Milewski
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (J.T.W.); (A.C.); (D.M.); (Y.Y.K.); (J.S.W.)
| | - Yong Y. Kim
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (J.T.W.); (A.C.); (D.M.); (Y.Y.K.); (J.S.W.)
| | - William D. Figg
- Clinical Pharmacology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA (N.C.)
| | - Elijah F. Edmondson
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (K.I.); (C.R.); (S.D.)
| | - Craig J. Thomas
- National Center for Advancing Translational Sciences, Rockville, MD 20850, USA; (X.Z.); (M.C.); (E.B.); (P.S.); (C.K.-T.); (K.M.W.); (C.M.); (Z.I.); (C.J.T.)
| | - Simone Difilippantonio
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (K.I.); (C.R.); (S.D.)
| | - Jun S. Wei
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (J.T.W.); (A.C.); (D.M.); (Y.Y.K.); (J.S.W.)
| | - Javed Khan
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (J.T.W.); (A.C.); (D.M.); (Y.Y.K.); (J.S.W.)
| |
Collapse
|
30
|
Ying H, Li ZQ, Li MP, Liu WC. Metabolism and senescence in the immune microenvironment of osteosarcoma: focus on new therapeutic strategies. Front Endocrinol (Lausanne) 2023; 14:1217669. [PMID: 37497349 PMCID: PMC10366376 DOI: 10.3389/fendo.2023.1217669] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 06/21/2023] [Indexed: 07/28/2023] Open
Abstract
Osteosarcoma is a highly aggressive and metastatic malignant tumor. It has the highest incidence of all malignant bone tumors and is one of the most common solid tumors in children and adolescents. Osteosarcoma tissues are often richly infiltrated with inflammatory cells, including tumor-associated macrophages, lymphocytes, and dendritic cells, forming a complex immune microenvironment. The expression of immune checkpoint molecules is also high in osteosarcoma tissues, which may be involved in the mechanism of anti-tumor immune escape. Metabolism and senescence are closely related to the immune microenvironment, and disturbances in metabolism and senescence may have important effects on the immune microenvironment, thereby affecting immune cell function and immune responses. Metabolic modulation and anti-senescence therapy are gaining the attention of researchers as emerging immunotherapeutic strategies for tumors. Through an in-depth study of the interconnection of metabolism and anti- senescence in the tumor immune microenvironment and its regulatory mechanism on immune cell function and immune response, more precise therapeutic strategies can be developed. Combined with the screening and application of biomarkers, personalized treatment can be achieved to improve therapeutic efficacy and provide a scientific basis for clinical decision-making. Metabolic modulation and anti- senescence therapy can also be combined with other immunotherapy approaches, such as immune checkpoint inhibitors and tumor vaccines, to form a multi-level and multi-dimensional immunotherapy strategy, thus further enhancing the effect of immunotherapy. Multidisciplinary cooperation and integrated treatment can optimize the treatment plan and maximize the survival rate and quality of life of patients. Future research and clinical practice will further advance this field, promising more effective treatment options for patients with osteosarcoma. In this review, we reviewed metabolic and senescence characteristics in the immune microenvironment of osteosarcoma and related immunotherapies, and provide a reference for development of more personalized and effective therapeutic strategies.
Collapse
Affiliation(s)
- Hui Ying
- Department of Emergency Trauma Surgery, Ganzhou People’s Hospital, Ganzhou, China
- Department of Spine Surgery, Ganzhou People’s Hospital, Ganzhou, China
| | - Zhi-Qiang Li
- Department of Emergency Trauma Surgery, Ganzhou People’s Hospital, Ganzhou, China
- Department of Spine Surgery, Ganzhou People’s Hospital, Ganzhou, China
| | - Meng-Pan Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wen-Cai Liu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
31
|
Nabbi A, Danesh A, Espin-Garcia O, Pedersen S, Wellum J, Fu LH, Paulson JN, Geoerger B, Marshall LV, Trippett T, Rossato G, Pugh TJ, Hutchinson KE. Multimodal immunogenomic biomarker analysis of tumors from pediatric patients enrolled to a phase 1-2 study of single-agent atezolizumab. NATURE CANCER 2023; 4:502-515. [PMID: 37038005 PMCID: PMC10132976 DOI: 10.1038/s43018-023-00534-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 02/24/2023] [Indexed: 04/12/2023]
Abstract
We report herein an exploratory biomarker analysis of refractory tumors collected from pediatric patients before atezolizumab therapy (iMATRIX-atezolizumab, NCT02541604 ). Elevated levels of CD8+ T cells and PD-L1 were associated with progression-free survival and a diverse baseline infiltrating T-cell receptor repertoire was prognostic. Differential gene expression analysis revealed elevated expression of CALCA (preprocalcitonin) and CCDC183 (highly expressed in testes) in patients who experienced clinical activity, suggesting that tumor neoantigens from these genes may contribute to immune response. In patients who experienced partial response or stable disease, elevated Igα2 expression correlated with T- and B-cell infiltration, suggesting that tertiary lymphoid structures existed in these patients' tumors. Consensus gene co-expression network analysis identified core cellular pathways that may play a role in antitumor immunity. Our study uncovers features associated with response to immune-checkpoint inhibition in pediatric patients with cancer and provides biological and translational insights to guide prospective biomarker profiling in future clinical trials.
Collapse
Affiliation(s)
- Arash Nabbi
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Arnavaz Danesh
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Osvaldo Espin-Garcia
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Epidemiology and Biostatistics, Western University, London, Ontario, Canada
- Dalla Lana School of Public Health and Department of Statistical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Stephanie Pedersen
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Johanna Wellum
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Lingyan Helen Fu
- Clinical Biomarker Operations, Product Development Oncology, Genentech, South San Francisco, CA, USA
| | - Joseph N Paulson
- Department of Biostatistics, Product Development, Genentech, South San Francisco, CA, USA
| | - Birgit Geoerger
- Gustave Roussy Cancer Centre, Department of Pediatric and Adolescent Oncology, INSERM U1015, Université Paris-Saclay, Villejuif, France
| | - Lynley V Marshall
- The Royal Marsden NHS Foundation Trust and the Institute of Cancer Research, London, UK
| | - Tanya Trippett
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gianluca Rossato
- Product Development Clinical Oncology, F. Hoffmann-La Roche, Basel, Switzerland
| | - Trevor J Pugh
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada.
| | | |
Collapse
|
32
|
Tian H, Cao J, Li B, Nice EC, Mao H, Zhang Y, Huang C. Managing the immune microenvironment of osteosarcoma: the outlook for osteosarcoma treatment. Bone Res 2023; 11:11. [PMID: 36849442 PMCID: PMC9971189 DOI: 10.1038/s41413-023-00246-z] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/17/2022] [Accepted: 12/29/2022] [Indexed: 03/01/2023] Open
Abstract
Osteosarcoma, with poor survival after metastasis, is considered the most common primary bone cancer in adolescents. Notwithstanding the efforts of researchers, its five-year survival rate has only shown limited improvement, suggesting that existing therapeutic strategies are insufficient to meet clinical needs. Notably, immunotherapy has shown certain advantages over traditional tumor treatments in inhibiting metastasis. Therefore, managing the immune microenvironment in osteosarcoma can provide novel and valuable insight into the multifaceted mechanisms underlying the heterogeneity and progression of the disease. Additionally, given the advances in nanomedicine, there exist many advanced nanoplatforms for enhanced osteosarcoma immunotherapy with satisfactory physiochemical characteristics. Here, we review the classification, characteristics, and functions of the key components of the immune microenvironment in osteosarcoma. This review also emphasizes the application, progress, and prospects of osteosarcoma immunotherapy and discusses several nanomedicine-based options to enhance the efficiency of osteosarcoma treatment. Furthermore, we examine the disadvantages of standard treatments and present future perspectives for osteosarcoma immunotherapy.
Collapse
Affiliation(s)
- Hailong Tian
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041 China
| | - Jiangjun Cao
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041 China
| | - Bowen Li
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041 China
| | - Edouard C. Nice
- grid.1002.30000 0004 1936 7857Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800 Australia
| | - Haijiao Mao
- Department of Orthopaedic Surgery, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang, 315020, People's Republic of China.
| | - Yi Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
33
|
Timpanaro A, Piccand C, Uldry AC, Bode PK, Dzhumashev D, Sala R, Heller M, Rössler J, Bernasconi M. Surfaceome Profiling of Cell Lines and Patient-Derived Xenografts Confirm FGFR4, NCAM1, CD276, and Highlight AGRL2, JAM3, and L1CAM as Surface Targets for Rhabdomyosarcoma. Int J Mol Sci 2023; 24:2601. [PMID: 36768928 PMCID: PMC9917031 DOI: 10.3390/ijms24032601] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/17/2023] [Accepted: 01/27/2023] [Indexed: 02/03/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma in children. The prognosis for patients with high-grade and metastatic disease is still very poor, and survivors are burdened with long-lasting side effects. Therefore, more effective and less toxic therapies are needed. Surface proteins are ideal targets for antibody-based therapies, like bispecific antibodies, antibody-drug conjugates, or chimeric antigen receptor (CAR) T-cells. Specific surface targets for RMS are scarce. Here, we performed a surfaceome profiling based on differential centrifugation enrichment of surface/membrane proteins and detection by LC-MS on six fusion-positive (FP) RMS cell lines, five fusion-negative (FN) RMS cell lines, and three RMS patient-derived xenografts (PDXs). A total of 699 proteins were detected in the three RMS groups. Ranking based on expression levels and comparison to expression in normal MRC-5 fibroblasts and myoblasts, followed by statistical analysis, highlighted known RMS targets such as FGFR4, NCAM1, and CD276/B7-H3, and revealed AGRL2, JAM3, MEGF10, GPC4, CADM2, as potential targets for immunotherapies of RMS. L1CAM expression was investigated in RMS tissues, and strong L1CAM expression was observed in more than 80% of alveolar RMS tumors, making it a practicable target for antibody-based therapies of alveolar RMS.
Collapse
Affiliation(s)
- Andrea Timpanaro
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Translational Cancer Research, Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Caroline Piccand
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Translational Cancer Research, Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Anne-Christine Uldry
- Proteomics & Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Peter Karl Bode
- Department of Pathology and Molecular Pathology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Dzhangar Dzhumashev
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Translational Cancer Research, Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Rita Sala
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Translational Cancer Research, Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Manfred Heller
- Proteomics & Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Jochen Rössler
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Translational Cancer Research, Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Michele Bernasconi
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Translational Cancer Research, Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, University of Zurich, 3032 Zurich, Switzerland
| |
Collapse
|
34
|
Hattinger CM, Salaroglio IC, Fantoni L, Godel M, Casotti C, Kopecka J, Scotlandi K, Ibrahim T, Riganti C, Serra M. Strategies to Overcome Resistance to Immune-Based Therapies in Osteosarcoma. Int J Mol Sci 2023; 24:ijms24010799. [PMID: 36614241 PMCID: PMC9821333 DOI: 10.3390/ijms24010799] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/14/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
Improving the prognosis and cure rate of HGOSs (high-grade osteosarcomas) is an absolute need. Immune-based treatment approaches have been increasingly taken into consideration, in particular for metastatic, relapsed and refractory HGOS patients, to ameliorate the clinical results currently achieved. This review is intended to give an overview on the immunotherapeutic treatments targeting, counteracting or exploiting the different immune cell compartments that are present in the HGOS tumor microenvironment. The principle at the basis of these strategies and the possible mechanisms that HGOS cells may use to escape these treatments are presented and discussed. Finally, a list of the currently ongoing immune-based trials in HGOS is provided, together with the results that have been obtained in recently completed clinical studies. The different strategies that are presently under investigation, which are generally aimed at abrogating the immune evasion of HGOS cells, will hopefully help to indicate new treatment protocols, leading to an improvement in the prognosis of patients with this tumor.
Collapse
Affiliation(s)
- Claudia Maria Hattinger
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | | | - Leonardo Fantoni
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy
| | - Martina Godel
- Department of Oncology, University of Torino, Via Santena 5/bis, 10126 Torino, Italy
| | - Chiara Casotti
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy
| | - Joanna Kopecka
- Department of Oncology, University of Torino, Via Santena 5/bis, 10126 Torino, Italy
| | - Katia Scotlandi
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Toni Ibrahim
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, Via Santena 5/bis, 10126 Torino, Italy
- Correspondence: (C.R.); (M.S.)
| | - Massimo Serra
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
- Correspondence: (C.R.); (M.S.)
| |
Collapse
|
35
|
Current State of Immunotherapy and Mechanisms of Immune Evasion in Ewing Sarcoma and Osteosarcoma. Cancers (Basel) 2022; 15:cancers15010272. [PMID: 36612267 PMCID: PMC9818129 DOI: 10.3390/cancers15010272] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
We argue here that in many ways, Ewing sarcoma (EwS) is a unique tumor entity and yet, it shares many commonalities with other immunologically cold solid malignancies. From the historical perspective, EwS, osteosarcoma (OS) and other bone and soft-tissue sarcomas were the first types of tumors treated with the immunotherapy approach: more than 100 years ago American surgeon William B. Coley injected his patients with a mixture of heat-inactivated bacteria, achieving survival rates apparently higher than with surgery alone. In contrast to OS which exhibits recurrent somatic copy-number alterations, EwS possesses one of the lowest mutation rates among cancers, being driven by a single oncogenic fusion protein, most frequently EWS-FLI1. In spite these differences, both EwS and OS are allied with immune tolerance and low immunogenicity. We discuss here the potential mechanisms of immune escape in these tumors, including low representation of tumor-specific antigens, low expression levels of MHC-I antigen-presenting molecules, accumulation of immunosuppressive M2 macrophages and myeloid proinflammatory cells, and release of extracellular vesicles (EVs) which are capable of reprogramming host cells in the tumor microenvironment and systemic circulation. We also discuss the vulnerabilities of EwS and OS and potential novel strategies for their targeting.
Collapse
|
36
|
Xue B, von Heyking K, Gassmann H, Poorebrahim M, Thiede M, Schober K, Mautner J, Hauer J, Ruland J, Busch DH, Thiel U, Burdach SEG. T Cells Directed against the Metastatic Driver Chondromodulin-1 in Ewing Sarcoma: Comparative Engineering with CRISPR/Cas9 vs. Retroviral Gene Transfer for Adoptive Transfer. Cancers (Basel) 2022; 14:cancers14225485. [PMID: 36428578 PMCID: PMC9688113 DOI: 10.3390/cancers14225485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/31/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022] Open
Abstract
Ewing sarcoma (EwS) is a highly malignant sarcoma of bone and soft tissue with early metastatic spread and an age peak in early puberty. The prognosis in advanced stages is still dismal, and the long-term effects of established therapies are severe. Efficacious targeted therapies are urgently needed. Our previous work has provided preliminary safety and efficacy data utilizing T cell receptor (TCR) transgenic T cells, generated by retroviral gene transfer, targeting HLA-restricted peptides on the tumor cell derived from metastatic drivers. Here, we compared T cells engineered with either CRISPR/Cas9 or retroviral gene transfer. Firstly, we confirmed the feasibility of the orthotopic replacement of the endogenous TCR by CRISPR/Cas9 with a TCR targeting our canonical metastatic driver chondromodulin-1 (CHM1). CRISPR/Cas9-engineered T cell products specifically recognized and killed HLA-A*02:01+ EwS cell lines. The efficiency of retroviral transduction was higher compared to CRISPR/Cas9 gene editing. Both engineered T cell products specifically recognized tumor cells and elicited cytotoxicity, with CRISPR/Cas9 engineered T cells providing prolonged cytotoxic activity. In conclusion, T cells engineered with CRISPR/Cas9 could be feasible for immunotherapy of EwS and may have the advantage of more prolonged cytotoxic activity, as compared to T cells engineered with retroviral gene transfer.
Collapse
Affiliation(s)
- Busheng Xue
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, School of Medicine, Technical University of Munich, 80804 Munich, Germany
| | - Kristina von Heyking
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, School of Medicine, Technical University of Munich, 80804 Munich, Germany
| | - Hendrik Gassmann
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, School of Medicine, Technical University of Munich, 80804 Munich, Germany
| | - Mansour Poorebrahim
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, School of Medicine, Technical University of Munich, 80804 Munich, Germany
| | - Melanie Thiede
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, School of Medicine, Technical University of Munich, 80804 Munich, Germany
| | - Kilian Schober
- Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich, 81674 Munich, Germany
| | - Josef Mautner
- Department of Gene Vectors, Helmholtz Centre Munich, 81377 Munich, Germany
- DZIF, German Center for Infection Research, Partner Site Munich, Germany Institute of Clinical, 81675 Munich, Germany
| | - Julia Hauer
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, School of Medicine, Technical University of Munich, 80804 Munich, Germany
- Munich Childhood Health Alliance (CHANCE) e.V, 80337 Munich, Germany
| | - Jürgen Ruland
- DZIF, German Center for Infection Research, Partner Site Munich, Germany Institute of Clinical, 81675 Munich, Germany
- DKTK German Cancer Consortium, Partner Site Munich, 81675 Munich, Germany
- Institute of Chemistry and Pathobiochemistry, TUM School of Medicine, Technical University of Munich, 81675 Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), 81675 Munich, Germany
| | - Dirk H. Busch
- Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich, 81674 Munich, Germany
- DZIF, German Center for Infection Research, Partner Site Munich, Germany Institute of Clinical, 81675 Munich, Germany
- Munich Childhood Health Alliance (CHANCE) e.V, 80337 Munich, Germany
| | - Uwe Thiel
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, School of Medicine, Technical University of Munich, 80804 Munich, Germany
- Munich Childhood Health Alliance (CHANCE) e.V, 80337 Munich, Germany
- Correspondence: (U.T.); (S.E.G.B.)
| | - Stefan E. G. Burdach
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, School of Medicine, Technical University of Munich, 80804 Munich, Germany
- Munich Childhood Health Alliance (CHANCE) e.V, 80337 Munich, Germany
- DKTK German Cancer Consortium, Partner Site Munich, 81675 Munich, Germany
- Translational Pediatric Cancer Research-Institute of Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- Department of Molecular Oncology, British Columbia Cancer Research Centre and Academy of Translational Medicine, University of British Columbia, Vancouver, BC V5Z 1L3, Canada
- Correspondence: (U.T.); (S.E.G.B.)
| |
Collapse
|
37
|
Kaczorowski M, Chłopek M, Kruczak A, Ryś J, Lasota J, Miettinen M. PRAME Expression in Cancer. A Systematic Immunohistochemical Study of >5800 Epithelial and Nonepithelial Tumors. Am J Surg Pathol 2022; 46:1467-1476. [PMID: 35973038 PMCID: PMC9588667 DOI: 10.1097/pas.0000000000001944] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Preferentially expressed antigen in melanoma (PRAME) is considered a useful marker in the differential diagnosis between malignant melanoma and its melanocytic mimics. Recently PRAME expression was documented in nonmelanocytic tumors, but much of the data are based on mRNA studies. This investigation evaluated PRAME expression in the spectrum of normal tissues and >5800 human tumors using immunohistochemistry and EP461 monoclonal antibody. In normal tissues, PRAME was expressed in the testis and proliferative endometrium. In tumors, PRAME was variably expressed in malignancies of different lineages. Among epithelial tumors, >50% of PRAME-positive lesions were found among endometrial carcinomas (82%), uterine serous carcinomas (82%), uterine carcinosarcomas (60%), ovarian clear cell carcinomas (90%), ovarian serous carcinomas (63%), adenoid cystic carcinomas (81%), seminomas (78%), thymic carcinomas (75%), and basal cell carcinomas (62%). In mesenchymal and neuroectodermal malignancies, PRAME was frequently expressed in synovial sarcoma (71%), myxoid liposarcoma (76%), neuroblastoma (61%) and metastatic melanoma (87%). Also, PRAME was consistently expressed in 4 melanomas that lacked all melanoma markers including S100 protein and SOX10 but harbored typical for melanoma BRAF or NRAS driver mutations. However, strong and diffuse PRAME immunoreactivity was seen in many types of nonmelanocytic poorly differentiated carcinomas and sarcomas. Based on this study, PRAME is a relatively unspecific immunohistochemical marker, which limits its use in diagnostic surgical pathology. However, immunohistochemistry is a reliable and unexpensive method useful in detecting PRAME-positive malignancies for potential immunotherapy.
Collapse
Affiliation(s)
- Maciej Kaczorowski
- Laboratory of Pathology, National Cancer Institute, Bethesda, Maryland, USA
- Department of Clinical and Experimental Pathology, Wroclaw Medical University, Wrocław, Poland
| | - Małgorzata Chłopek
- Laboratory of Pathology, National Cancer Institute, Bethesda, Maryland, USA
| | - Anna Kruczak
- Department of Tumor Pathology, Maria Skłodowska-Curie National Research Institute of Oncology, Cracow Branch, Poland
| | - Janusz Ryś
- Department of Tumor Pathology, Maria Skłodowska-Curie National Research Institute of Oncology, Cracow Branch, Poland
| | - Jerzy Lasota
- Laboratory of Pathology, National Cancer Institute, Bethesda, Maryland, USA
| | - Markku Miettinen
- Laboratory of Pathology, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
38
|
An Analysis of Transcriptomic Burden Identifies Biological Progression Roadmaps for Hematological Malignancies and Solid Tumors. Biomedicines 2022; 10:biomedicines10112720. [DOI: 10.3390/biomedicines10112720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Abstract
Biological paths of tumor progression are difficult to predict without time-series data. Using median shift and abacus transformation in the analysis of RNA sequencing data sets, natural patient stratifications were found based on their transcriptomic burden (TcB). Using gene-behavior analysis, TcB groups were evaluated further to discover biological courses of tumor progression. We found that solid tumors and hematological malignancies (n = 4179) share conserved biological patterns, and biological network complexity decreases at increasing TcB levels. An analysis of gene expression datasets including pediatric leukemia patients revealed TcB patterns with biological directionality and survival implications. A prospective interventional study with PI3K targeted therapy in canine lymphomas proved that directional biological responses are dynamic. To conclude, TcB-enriched biological mechanisms detected the existence of biological trajectories within tumors. Using this prognostic informative novel informatics method, which can be applied to tumor transcriptomes and progressive diseases inspires the design of progression-specific therapeutic approaches.
Collapse
|
39
|
Dzhumashev D, Timpanaro A, Ali S, De Micheli AJ, Mamchaoui K, Cascone I, Rössler J, Bernasconi M. Quantum Dot-Based Screening Identifies F3 Peptide and Reveals Cell Surface Nucleolin as a Therapeutic Target for Rhabdomyosarcoma. Cancers (Basel) 2022; 14:5048. [PMID: 36291832 PMCID: PMC9600270 DOI: 10.3390/cancers14205048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
Active drug delivery by tumor-targeting peptides is a promising approach to improve existing therapies for rhabdomyosarcoma (RMS), by increasing the therapeutic effect and decreasing the systemic toxicity, e.g., by drug-loaded peptide-targeted nanoparticles. Here, we tested 20 different tumor-targeting peptides for their ability to bind to two RMS cell lines, Rh30 and RD, using quantum dots Streptavidin and biotin-peptides conjugates as a model for nanoparticles. Four peptides revealed a very strong binding to RMS cells: NCAM-1-targeting NTP peptide, nucleolin-targeting F3 peptide, and two Furin-targeting peptides, TmR and shTmR. F3 peptide showed the strongest binding to all RMS cell lines tested, low binding to normal control myoblasts and fibroblasts, and efficient internalization into RMS cells demonstrated by the cytoplasmic delivery of the Saporin toxin. The expression of the nucleophosphoprotein nucleolin, the target of F3, on the surface of RMS cell lines was validated by competition with the natural ligand lactoferrin, by colocalization with the nucleolin-binding aptamer AS1411, and by the marked sensitivity of RMS cell lines to the growth inhibitory nucleolin-binding N6L pseudopeptide. Taken together, our results indicate that nucleolin-targeting by F3 peptide represents a potential therapeutic approach for RMS.
Collapse
Affiliation(s)
- Dzhangar Dzhumashev
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Andrea Timpanaro
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Safa Ali
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Andrea J. De Micheli
- Department of Oncology, University Children’s Hospital Zurich, 8032 Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, University of Zurich, 3032 Zurich, Switzerland
| | - Kamel Mamchaoui
- Centre de Recherche en Myologie, Institut de Myologie, INSERM, Sorbonne Université, F-75013 Paris, France
| | - Ilaria Cascone
- IMRB, INSERM, University Paris Est Creteil, 94010 Creteil, France
- AP-HP, Groupe Hospitalo-Universitaire Chenevier Mondor, Centre d’Investigation Clinique Biothérapie, 94010 Créteil, France
| | - Jochen Rössler
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Michele Bernasconi
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, University of Zurich, 3032 Zurich, Switzerland
| |
Collapse
|
40
|
Abstract
Undifferentiated small round cell sarcomas (SRCSs) of bone and soft tissue comprise a heterogeneous group of highly aggressive tumours associated with a poor prognosis, especially in metastatic disease. SRCS entities mainly occur in the third decade of life and can exhibit striking disparities regarding preferentially affected sex and tumour localization. SRCSs comprise new entities defined by specific genetic abnormalities, namely EWSR1-non-ETS fusions, CIC-rearrangements or BCOR genetic alterations, as well as EWSR1-ETS fusions in the prototypic SRCS Ewing sarcoma. These gene fusions mainly encode aberrant oncogenic transcription factors that massively rewire the transcriptome and epigenome of the as yet unknown cell or cells of origin. Additional mutations or copy number variants are rare at diagnosis and, depending on the tumour entity, may involve TP53, CDKN2A and others. Histologically, these lesions consist of small round cells expressing variable levels of CD99 and specific marker proteins, including cyclin B3, ETV4, WT1, NKX3-1 and aggrecan, depending on the entity. Besides locoregional treatment that should follow standard protocols for sarcoma management, (neo)adjuvant treatment is as yet ill-defined but generally follows that of Ewing sarcoma and is associated with adverse effects that might compromise quality of life. Emerging studies on the molecular mechanisms of SRCSs and the development of genetically engineered animal models hold promise for improvements in early detection, disease monitoring, treatment-related toxicity, overall survival and quality of life.
Collapse
|
41
|
Sparber-Sauer M. The future of rhabdomyosarcoma trials. THE LANCET. CHILD & ADOLESCENT HEALTH 2022; 6:510-511. [PMID: 35690070 DOI: 10.1016/s2352-4642(22)00132-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 04/22/2022] [Indexed: 06/15/2023]
Affiliation(s)
- Monika Sparber-Sauer
- Hospital of the State Capital Stuttgart gKAöR, Center for Pediatric, Adolescent and Women's Medicine, Central location (Olgahospital), D-70174 Stuttgart, Germany.
| |
Collapse
|
42
|
Thorner PS, Chong AS, Nadaf J, Benlimame N, Marrano P, Chami R, Fu L, Foulkes WD. PRAME protein expression in DICER1-related tumours. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2022; 8:294-304. [PMID: 35297207 PMCID: PMC8977278 DOI: 10.1002/cjp2.264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/28/2022] [Accepted: 01/30/2022] [Indexed: 11/09/2022]
Abstract
DICER1 syndrome is an autosomal dominant tumour predisposition syndrome usually affecting persons under 30 years of age. Many of the associated benign and malignant lesions occur almost exclusively in DICER1 syndrome. One such tumour, pituitary blastoma (pitB), overexpresses PRAME 500x above control levels. PRAME (PReferentially expressed Antigen in MElanoma) is expressed in malignancies that are not DICER1-related (e.g. melanoma). To address whether PRAME expression is part of the DICER1 phenotype, or simply a feature of pitB, a series of 75 DICER1-mutated specimens and 33 non-mutated specimens was surveyed using immunohistochemistry for PRAME, together with EZH2, which complexes with PRAME. In DICER1-mutated specimens, positive staining for PRAME was only seen in malignant tumours; 7 of 11 histological types and 34/62 individual tumours were positive, while non-tumourous lesions were always negative. Pleuropulmonary blastoma (PPB) showed a continuum in staining, with type I lesions being PRAME negative (n = 7) but all type II and type III lesions PRAME positive (n = 7). Similarly, cystic nephroma (CN) was negative (n = 8), with anaplastic sarcoma of the kidney being positive (n = 2). However, one atypical CN with mesenchymal cell proliferation was PRAME-positive. Embryonal rhabdomyosarcoma (RMS) with DICER1 pathogenic variants (PVs) was positive for PRAME (5/6), but the same tumour type without DICER1 PVs was also positive (9/15). Staining for EZH2 corresponded to that seen with PRAME, validating the latter. This study leads us to conclude that (1) PRAME expression occurs in two-thirds of DICER1-related malignancies; (2) PRAME may be a marker for the progression that certain DICER1-related lesions are thought to undergo, such as PPB and CN; and (3) PRAME expression in some tumours, such as RMS, appears to be an intrinsic feature of the tumour, rather than specifically related to DICER1 PVs. Therapy directed against PRAME may offer novel treatment options in patients with the DICER1 syndrome.
Collapse
Affiliation(s)
- Paul S Thorner
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Anne-Sophie Chong
- Department of Human Genetics, McGill University, Montreal, QC, Canada.,Cancer Axis, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada.,Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Javad Nadaf
- Department of Human Genetics, McGill University, Montreal, QC, Canada.,Cancer Axis, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
| | - Naciba Benlimame
- Research Pathology Facility, Lady Davis Institute, Jewish General Hospital, Montreal, QC, Canada
| | - Paula Marrano
- Division of Pathology, Hospital for Sick Children, Toronto, ON, Canada
| | - Rose Chami
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Division of Pathology, Hospital for Sick Children, Toronto, ON, Canada
| | - Lili Fu
- Department of Pathology, McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - William D Foulkes
- Department of Human Genetics, McGill University, Montreal, QC, Canada.,Cancer Axis, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada.,Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada
| |
Collapse
|