1
|
Souto EP, Gong P, Landua JD, Rajaram Srinivasan R, Ganesan A, Dobrolecki LE, Purdy SC, Pan X, Zeosky M, Chung A, Yi SS, Ford HL, Lewis MT. Lineage Tracing and Single-Cell RNA Sequencing Reveal a Common Transcriptional State in Breast Cancer Tumor-Initiating Cells Characterized by IFN/STAT1 Activity. Cancer Res 2025; 85:1390-1409. [PMID: 40230213 PMCID: PMC11997551 DOI: 10.1158/0008-5472.can-23-4022] [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: 12/19/2023] [Revised: 09/03/2024] [Accepted: 01/31/2025] [Indexed: 04/16/2025]
Abstract
A tumor cell subpopulation of tumor-initiating cells (TIC) or "cancer stem cells" is associated with therapeutic resistance, as well as both local and distant recurrences. Signal transducer and activator of transcription (STAT) activity is elevated in TICs in claudin-low models of human triple-negative breast cancer, which enables enrichment of TICs using a STAT-responsive reporter. Lineage tracing of TICs as they undergo cell state changes could enable a better understanding of the molecular phenotypes of TIC and uncover strategies to selectively target TICs. In this study, we developed a STAT-responsive lineage-tracing system and used it in conjunction with the original reporter to enrich for cells with enhanced mammosphere-forming potential. This approach was able to detect TICs in some, but not all, basal-like triple-negative breast cancer xenograft models, indicating that STAT signaling has both TIC-related and TIC-independent functions. Single-cell RNA sequencing (RNA-seq) of reporter-tagged xenografts and clinical samples identified a common IFN/STAT1-associated transcriptional state in TICs that was previously linked to inflammation and macrophage differentiation. Surprisingly, most of the identified genes were not present in previously published TIC signatures derived using bulk RNA-seq. Finally, bone marrow stromal cell antigen-2 was identified as a cell surface marker of this state that functionally regulated TIC frequency. These results suggest that TICs may exploit the IFN/STAT1 signaling axis to promote their activity and that targeting this pathway may help eliminate TICs. Significance: Coupling single-cell transcriptomics with tumor-initiating cell enrichment identified IFN response gene expression not previously reported in bulk RNA-sequencing-derived signatures and proposed IFN/STAT1 signaling as a candidate therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Eric P. Souto
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Ping Gong
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - John D. Landua
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | | | - Abhinaya Ganesan
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Lacey E. Dobrolecki
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Stephen C. Purdy
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (UC-AMC), Aurora, Colorado
- Pharmacology Graduate Program, UC-AMC, Aurora, Colorado
- University of Colorado Cancer Center, UC-AMC, Aurora, Colorado
| | - Xingxin Pan
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas
| | - Michael Zeosky
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas
| | - Anna Chung
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas
| | - S. Stephen Yi
- Livestrong Cancer Institutes, The University of Texas at Austin, Austin, Texas
| | - Heide L. Ford
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (UC-AMC), Aurora, Colorado
- Pharmacology Graduate Program, UC-AMC, Aurora, Colorado
- University of Colorado Cancer Center, UC-AMC, Aurora, Colorado
| | - Michael T. Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, Texas
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Department of Radiology, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
2
|
Mendes RV, Ribeiro JM, Gouveia H, Rebelo de Almeida C, Castillo-Martin M, Brito MJ, Canas-Marques R, Batista E, Alves C, Sousa B, Gouveia P, Ferreira MG, Cardoso MJ, Cardoso F, Fior R. Zebrafish Avatar testing preclinical study predicts chemotherapy response in breast cancer. NPJ Precis Oncol 2025; 9:94. [PMID: 40169839 PMCID: PMC11961725 DOI: 10.1038/s41698-025-00882-0] [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: 10/22/2024] [Accepted: 03/15/2025] [Indexed: 04/03/2025] Open
Abstract
Chemotherapy remains the mainstay in most high-risk breast cancer (BC) settings, with several equivalent options of treatment. However, the efficacy of each treatment varies between patients and there is currently no test to determine which option will be the most effective for each individual patient. Here, we developed a fast in-vivo test for BC therapy screening: the zebrafish patient-derived-xenograft model (zAvatars), where in-vivo results can be obtained in just 10 days. To determine the predictive value of the BC zAvatars we performed a preclinical study, where zAvatars were treated with the same therapy as the donor-patient and their response to therapy was compared. Our data show a 100% concordance (18 out of 18) between the zAvatar-test and the corresponding patient's clinical response to treatment. Altogether, our results suggest that the zAvatar model constitutes a promising in-vivo assay to optimize cancer treatments in a truly personalized manner.
Collapse
Affiliation(s)
- Raquel V Mendes
- Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| | - Joana M Ribeiro
- Gustave Roussy, Département de Médecine Oncologique, Villejuif, France
| | - Helena Gouveia
- Breast Unit, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal
| | | | - Mireia Castillo-Martin
- Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
- Service of Pathology, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal
| | - Maria José Brito
- Service of Pathology, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal
| | - Rita Canas-Marques
- Service of Pathology, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal
| | - Eva Batista
- Breast Unit, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal
| | - Celeste Alves
- Breast Unit, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal
| | - Berta Sousa
- Breast Unit, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal
| | - Pedro Gouveia
- Breast Unit, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal
| | - Miguel Godinho Ferreira
- Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR7284 INSERM U1081, Université Côte d'Azur, 06107, Nice, France
| | - Maria João Cardoso
- Breast Unit, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal
- Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Fatima Cardoso
- Breast Unit, Champalimaud Clinical Center, Champalimaud Foundation, Lisbon, Portugal
| | - Rita Fior
- Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal.
| |
Collapse
|
3
|
Arriojas A, Baek LM, Berner MJ, Zhurkevich A, Hinton AO, Meyer MD, Dobrolecki LE, Lewis MT, Zarringhalam K, Echeverria GV. Artificial intelligence-enabled automated analysis of transmission electron micrographs to evaluate chemotherapy impact on mitochondrial morphology in triple negative breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.19.635300. [PMID: 40027627 PMCID: PMC11870520 DOI: 10.1101/2025.02.19.635300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Advancements in transmission electron microscopy (TEM) have enabled in-depth studies of biological specimens, offering new avenues to large-scale imaging experiments with subcellular resolution. Mitochondrial structure is of growing interest in cancer biology due to its crucial role in regulating the multi-faceted functions of mitochondria. We and others have established the crucial role of mitochondria in triple-negative breast cancer (TNBC), an aggressive subtype of breast cancer with limited therapeutic options. Building upon our previous work demonstrating the regulatory role of mitochondrial structure dynamics in metabolic adaptation and survival of chemotherapy-refractory TNBC cells, we sought to extend those findings to a large-scale analysis of transmission electron micrographs. Here we present a UNet artificial intelligence (AI) model for automatic annotation and assessment of mitochondrial morphology and feature quantification. Our model is trained on 11,039 manually annotated mitochondria across 125 micrographs derived from a variety of orthotopic patient-derived xenograft (PDX) mouse model tumors and adherent cell cultures. The model achieves an F1 score of 0.85 on test micrographs at the pixel level. To validate the ability of our model to detect expected mitochondrial structural features, we utilized micrographs from mouse primary skeletal muscle cells genetically modified to lack Dynamin-related protein 1 (Drp1). The algorithm successfully detected a significant increase in mitochondrial elongation, in alignment with the well-established role of Drp1 as a driver of mitochondrial fission. Further, we subjected in vitro and in vivo TNBC models to conventional chemotherapy treatments commonly used for clinical management of TNBC, including doxorubicin, carboplatin, paclitaxel, and docetaxel (DTX). We found substantial within-sample heterogeneity of mitochondrial structure in both in vitro and in vivo TNBC models and observed a consistent reduction in mitochondrial elongation in DTX-treated specimens. We went on to compare mammary tumors and matched lung metastases in a highly metastatic PDX model of TNBC, uncovering significant reduction in mitochondrial length in metastatic lesions. Our large, curated dataset provides high statistical power to detect frequent chemotherapy-induced shifts in mitochondrial shapes and sizes in residual cells left behind after treatment. The successful application of our AI model to capture mitochondrial structure marks a step forward in high-throughput analysis of mitochondrial structures, enhancing our understanding of how morphological changes may relate to chemotherapy efficacy and mechanism of action. Finally, our large, manually curated electron micrograph dataset - now publicly available - serves as a unique gold-standard resource for developing, benchmarking, and applying computational models, while further advancing investigations into mitochondrial morphology and its impact on cancer biology.
Collapse
|
4
|
Wang D, Wang L, Zhang W, Xu K, Chen L, Guo Z, Wu K, Huang D, Zhao Y, Yao M, Zheng L, Ye C, Ran J, Zhou W, Liu X, Xu J. Extracellular vesicle-mediated gene therapy targets BRAF V600E-mutant colorectal cancer by inhibiting the MEK1/2-ERK1/2 pathway. J Nanobiotechnology 2025; 23:129. [PMID: 39979881 PMCID: PMC11843959 DOI: 10.1186/s12951-025-03205-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/05/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Patients with colorectal cancer (CRC) harboring BRAF mutation have a poor prognosis. The median survival time for patients with advanced BRAFV600E-mutant CRC is only approximately one year. Owing to the insensitivity to standard chemotherapy, there are still no effective and highly specific treatment strategies available in clinical practice for CRC patients with BRAF mutation. Therefore, targeting the BRAFV600E mutation site, researching and exploring novel targeted therapies are essential to improve the survival rate of patients with this CRC subtype. AIM This study aims to develop a precise therapeutic system for BRAFV600E CRC, based on the carrier properties of extracellular vesicles (EVs) and gene therapy targeting BRAFV600E. METHOD We first obtained engineered cells capable of stably producing EVs loaded with BRAFV600E nucleic acid drugs (siBRAFV600E). Next, BRAFV600E-mutant and wild-type CRC cell lines, as well as corresponding subcutaneous and metastasis models, were used to evaluate the therapeutic efficacy of EVs-siBRAFV600E and explored the mechanism. Notably, patient-derived xenograft (PDX) models, which share the same molecular characteristics, pathological features, and heterogeneity as patients do, were utilized to further explore the therapeutic efficacy and mechanisms. RESULT EVs-siBRAFV600E specifically inhibited BRAFV600E CRC but didn't affect BRAF wild-type CRC in vitro and vivo. EVs-siBRAFV600E exerts its therapeutic effect by regulating the MEK1/2-ERK1/2 pathway, and it has demonstrated excellent therapeutic efficacy in PDX models. CONCLUSION The therapeutic EVs we constructed are effective and specific for the BRAFV600E-mutant CRC. This study provides a novel strategy for the treatment of CRC patients with BRAFV600E mutation.
Collapse
Affiliation(s)
- Di Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
| | - Liwei Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
| | - Wei Zhang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, PR China
| | - Kaicheng Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
| | - Liang Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
| | - Ziye Guo
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
| | - Kaile Wu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
| | - Donghua Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
| | - Yubin Zhao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
| | - Minjun Yao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
| | - Liming Zheng
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
| | - Chenyi Ye
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
| | - Jisheng Ran
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, 310009, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China
| | - Wei Zhou
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, PR China.
| | - Xin Liu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, PR China.
| | - Jianbin Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, PR China.
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, 310009, PR China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China.
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310009, PR China.
| |
Collapse
|
5
|
Palczewski MB, Kuschman HP, Hoffman BM, Kathiresan V, Yang H, Glynn SA, Wilson DL, Kool ET, Montfort WR, Chang J, Petenkaya A, Chronis C, Cundari TR, Sappa S, Islam K, McVicar DW, Fan Y, Chen Q, Meerzaman D, Sierk M, Thomas DD. Nitric oxide inhibits ten-eleven translocation DNA demethylases to regulate 5mC and 5hmC across the genome. Nat Commun 2025; 16:1732. [PMID: 39966373 PMCID: PMC11836389 DOI: 10.1038/s41467-025-56928-1] [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: 04/01/2024] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
DNA methylation at cytosine bases (5-methylcytosine, 5mC) is a heritable epigenetic mark regulating gene expression. While enzymes that metabolize 5mC are well-characterized, endogenous signaling molecules that regulate DNA methylation machinery have not been described. We report that physiological nitric oxide (NO) concentrations reversibly inhibit the DNA demethylases TET and ALKBH2 by binding to the mononuclear non-heme iron atom forming a dinitrosyliron complex (DNIC) and preventing cosubstrates from binding. In cancer cells treated with exogenous NO, or endogenously synthesizing NO, 5mC and 5-hydroxymethylcytosine (5hmC) increase, with no changes in DNA methyltransferase activity. 5mC is also significantly increased in NO-producing patient-derived xenograft tumors from mice. Genome-wide methylome analysis of cells chronically treated with NO (10 days) shows enrichment of 5mC and 5hmC at gene-regulatory loci, correlating with altered expression of NO-regulated tumor-associated genes. Regulation of DNA methylation is distinctly different from canonical NO signaling and represents a unique epigenetic role for NO.
Collapse
Affiliation(s)
- Marianne B Palczewski
- Department of Pharmaceutical Sciences, University of Illinois Chicago, College of Pharmacy, Chicago, IL, USA
| | - Hannah Petraitis Kuschman
- Department of Pharmaceutical Sciences, University of Illinois Chicago, College of Pharmacy, Chicago, IL, USA
| | - Brian M Hoffman
- Department of Chemistry, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA
| | - Venkatesan Kathiresan
- Department of Chemistry, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA
| | - Hao Yang
- Department of Chemistry, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA
| | - Sharon A Glynn
- Discipline of Pathology, University of Galway, College of Medicine, Nursing and Health Sciences, School of Medicine, Galway, Ireland
| | - David L Wilson
- Department of Chemistry, Stanford University, School of Humanities and Sciences, Stanford, CA, USA
| | - Eric T Kool
- Department of Chemistry, Stanford University, School of Humanities and Sciences, Stanford, CA, USA
| | - William R Montfort
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, USA
| | - Jenny Chang
- Dr. Mary and Neal Cancer Center at Houston Methodist, Weill Cornell Medical College, Houston, NY, USA
| | - Aydolun Petenkaya
- Department of Biomedical Engineering, University of Illinois Chicago, College of Engineering, Chicago, IL, USA
| | - Constantinos Chronis
- Department of Biochemistry and Molecular Genetics, University of Illinois Chicago, College of Medicine, Chicago, IL, USA
| | - Thomas R Cundari
- Department of Chemistry, University of North Texas, Denton, TX, USA
| | - Sushma Sappa
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kabirul Islam
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel W McVicar
- Cancer Innovation Laboratory, National Cancer Institute, Center for Cancer Research, Frederick, MD, USA
| | - Yu Fan
- National Cancer Institute, Center for Biomedical Informatics and Information Technology, Bethesda, USA
| | - Qingrong Chen
- National Cancer Institute, Center for Biomedical Informatics and Information Technology, Bethesda, USA
| | - Daoud Meerzaman
- Cancer Innovation Laboratory, National Cancer Institute, Center for Cancer Research, Frederick, MD, USA
| | - Michael Sierk
- Cancer Innovation Laboratory, National Cancer Institute, Center for Cancer Research, Frederick, MD, USA
| | - Douglas D Thomas
- Department of Pharmaceutical Sciences, University of Illinois Chicago, College of Pharmacy, Chicago, IL, USA.
| |
Collapse
|
6
|
Abdulkareem NM, Bhat R, Castillo M, Jung SY, Vasaikar S, Nanda S, Ruiz A, Shea M, Cao W, Veeraraghavan J, Kim HY, Bawa-Khalfe T, Hussain T, Liu X, Gunaratne P, Schiff R, Trivedi MV. Interactions between ADGRF1 (GPR110) and extracellular matrix proteins govern its effects on tumorigenesis in HER2-positive breast cancer. Br J Pharmacol 2025. [PMID: 39965212 DOI: 10.1111/bph.17463] [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: 03/07/2024] [Revised: 12/22/2024] [Accepted: 12/28/2024] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND AND PURPOSE We and others have previously shown that ADGRF1, an adhesion G protein-coupled receptor, is overexpressed and associated with poor survival in many cancers, including human epidermal growth factor receptor-2 (HER2) breast cancer (BC). Also, we have reported the tumour-promoting function of ADGRF1 using preclinical models of HER2+ BC. In this study, we investigated the effect of ADGRF1 overexpression in an orthotopic in vivo model as well as downstream signalling of ADGRF1 in HER2+ BC. EXPERIMENTAL APPROACH We utilized a doxycycline (Dox)-induced ADGRF1 overexpression system in HER2+ BC cell lines and performed various in vitro and in vivo studies. Following ADGRF1 overexpression in the presence/absence of Matrigel, laminin-111 or collagen-IV, we performed the mammosphere assay to assess the tumorigenicity of breast epithelial cells, as well as cAMP/IP1 assays and RNA-sequencing, to understand the receptor function and pharmacology. We conducted cross-linking-aided immunoprecipitation and mass spectrometry to confirm the physical interaction between ADGRF1 and the extracellular matrix proteins present in Matrigel. KEY RESULTS We found that ADGRF1 switched from a tumour-promoting to tumour-suppressive function upon interaction with laminin-111. Interaction of ADGRF1 with laminin-111 resulted in inhibition of Gαs coupling and STAT3 phosphorylation, induction of senescence, increase in HER2 expression, and improvement of sensitivity to anti-HER2 drugs in HER2+ BC. CONCLUSIONS ADGRF1 switches from a tumour-promoting to tumour-suppressive function upon interaction with laminin-111, leading to improvements in sensitivity to anti-HER2 drugs. Leveraging ADGRF1 interactions with laminin-111 may allow the design of novel therapies against ADGRF1 in HER2+ BC.
Collapse
Affiliation(s)
- Noor Mazin Abdulkareem
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA
| | - Raksha Bhat
- Department of Pharmacy Practice and Translational Research, University of Houston, Houston, Texas, USA
| | - Micah Castillo
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Sung Yun Jung
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Suhas Vasaikar
- Clinical Biomarker and Diagnostics, Seagen, Bothell, Washington, USA
| | - Sarmistha Nanda
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
| | - Alexis Ruiz
- Department of Pharmacy Practice and Translational Research, University of Houston, Houston, Texas, USA
| | - Martin Shea
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
| | - Wangjia Cao
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA
| | - Jamunarani Veeraraghavan
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Hee-Yong Kim
- Laboratory of Molecular Signaling, National Institute of Alcohol Abuse and Alcoholism, Rockville, Maryland, USA
| | - Tasneem Bawa-Khalfe
- Center for Nuclear Receptors & Cell Signaling, Department of Biology & Biochemistry, University of Houston, Houston, Texas, USA
| | - Tahir Hussain
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA
| | - Xinli Liu
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA
| | - Preethi Gunaratne
- Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Rachel Schiff
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Meghana V Trivedi
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA
- Department of Pharmacy Practice and Translational Research, University of Houston, Houston, Texas, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
7
|
Shea A, Eyal-Lubling Y, Guerrero-Romero D, Manzano Garcia R, Greenwood W, O’Reilly M, Georgopoulou D, Callari M, Lerda G, Wix S, Giovannetti A, Masina R, Esmaeilishirazifard E, Cope W, Martin AG, Nagano A, Young L, Kupczak S, Cheng Y, Bardwell H, Provenzano E, Kane J, Lay J, Grybowicz L, McAdam K, Caldas C, Abraham J, Rueda OM, Bruna A. Modeling Drug Responses and Evolutionary Dynamics Using Patient-Derived Xenografts Reveals Precision Medicine Strategies for Triple-Negative Breast Cancer. Cancer Res 2025; 85:567-584. [PMID: 39514406 PMCID: PMC7617242 DOI: 10.1158/0008-5472.can-24-1703] [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: 05/22/2024] [Revised: 08/09/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
The intertumor and intratumor heterogeneity of triple-negative breast cancers, which is reflected in diverse drug responses, interplays with tumor evolution. In this study, we developed a preclinical experimental and analytical framework using patient-derived tumor xenografts (PDTX) from patients with treatment-naïve triple-negative breast cancers to test their predictive value in personalized cancer treatment approaches. Patients and their matched PDTXs exhibited concordant drug responses to neoadjuvant therapy using two trial designs and dosing schedules. This platform enabled analysis of nongenetic mechanisms involved in relapse dynamics. Treatment resulted in permanent phenotypic changes, with functional and therapeutic consequences. High-throughput drug screening methods in ex vivo PDTX cells revealed patient-specific drug response changes dependent on first-line therapy. This was validated in vivo, as exemplified by a change in olaparib sensitivity in tumors previously treated with clinically relevant cycles of standard-of-care chemotherapy. In summary, PDTXs provide a robust tool to test patient drug responses and therapeutic regimens and to model evolutionary trajectories. However, high intermodel variability and permanent nongenomic transcriptional changes constrain their use for personalized cancer therapy. This work highlights important considerations associated with preclinical drug response modeling and potential uses of the platform to identify efficacious and preferential sequential therapeutic regimens. Significance: Patient-derived tumor xenografts from treatment-naïve breast cancer samples can predict patient drug responses and model treatment-induced phenotypic and functional evolution, making them valuable preclinical tools.
Collapse
Affiliation(s)
- Abigail Shea
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
- Institute of Cancer Research, London, United Kingdom
| | - Yaniv Eyal-Lubling
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Daniel Guerrero-Romero
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Raquel Manzano Garcia
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Wendy Greenwood
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Martin O’Reilly
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Dimitra Georgopoulou
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Maurizio Callari
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
- Fondazione Michelangelo, Milan, Italy
| | - Giulia Lerda
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Sophia Wix
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Agnese Giovannetti
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
- Clinical Genomics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, Foggia, Italy
| | - Riccardo Masina
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Elham Esmaeilishirazifard
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Wei Cope
- Department of Histopathology, Cambridge University NHS Foundation Trust, Cambridge, United Kingdom
| | - Alistair G. Martin
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Ai Nagano
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Lisa Young
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Steven Kupczak
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Yi Cheng
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Helen Bardwell
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Elena Provenzano
- Department of Histopathology, Cambridge University NHS Foundation Trust, Cambridge, United Kingdom
- Cambridge NIH Biomedical Research Centre, Cambridge, United Kingdom
| | - Justine Kane
- Department of Oncology, Precision Breast Cancer Institute, University of Cambridge, Cambridge, United Kingdom
| | - Jonny Lay
- Department of Oncology, Precision Breast Cancer Institute, University of Cambridge, Cambridge, United Kingdom
| | - Louise Grybowicz
- Cambridge University NHS Foundation Trust, Cambridge, United Kingdom
| | - Karen McAdam
- Cambridge University NHS Foundation Trust, Cambridge, United Kingdom
| | - Carlos Caldas
- Department of Oncology, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Clinical Biochemistry, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Jean Abraham
- Department of Oncology, Precision Breast Cancer Institute, University of Cambridge, Cambridge, United Kingdom
| | - Oscar M. Rueda
- MRC-Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom
| | | |
Collapse
|
8
|
Pataccini G, Elia A, Sequeira G, Ambrosio L, Coianis M, Lamb CA, Rojas PA, Martínez Vázquez P, Burruchaga J, Spengler E, Vanzulli SI, Abba M, Lanari C. Steroid hormone receptors, exome sequencing and treatment responsiveness of breast cancer patient-derived xenografts originated in a South American country. Sci Rep 2025; 15:2415. [PMID: 39827242 PMCID: PMC11742900 DOI: 10.1038/s41598-025-86389-x] [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: 09/10/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
Breast cancer (BC) patient-derived xenografts (PDX) are relevant models for precision medicine. However, there are no collections derived from South American BC patients. Since ethnicity significantly impacts clinical outcomes, it is necessary to develop PDX models from different lineages. Our goals were to a) develop BC PDX from our population; b) characterize the expression of estrogen (ER), progesterone (PR), androgen (AR) and glucocorticoid (GR) receptors, basal and luminal cytokeratins, EGFR and HER2; c) identify PDX mutations; d) evaluate the response to treatments selected based on their biological and genetic features, and e) perform BC tissue cultures (BCTC) from PDX tissues and compare in vivo and ex vivo results. Surgical fragments were maintained in a culture medium and inoculated subcutaneously into untreated NSG female mice, or treated with estradiol pellets. Other fragments were fixed in formalin for diagnosis and immunohistochemistry, and a third piece was frozen at -80°C for molecular studies or whole exome-sequencing. Tumors were serially transplanted into NSG mice. Once the PDX was established, in vivo and ex vivo drug responses were evaluated. Eight PDX were established: two ER + [BC-AR685 (PR +) and BC-AR707 (PR-)], one from a triple-negative (TN) recurrence whose primary tumor was ER + (BC-AR485), one HER2 + (BC-AR474) and four TN primary tumors (BC-AR553, BC-AR546, BC-AR631 and BC-AR687). BC-AR685 had higher levels of PR isoform A than isoform B and was sensitive to mifepristone, tamoxifen, and palbociclib. BC-AR707 was inhibited by tamoxifen and testosterone. BC-AR474 was inhibited by trastuzumab and trastuzumab emtansine. BC-AR485 was sensitive to doxorubicin and resistant to paclitaxel in vivo and ex vivo. BC-AR687 carried a PIK3CA (C420R) mutation and was sensitive to alpelisib and mTOR inhibitors. All PDX expressed AR with varying intensities. GR and AR were co-expressed in the ER + tumors and in 3 TN PDX. We report the first PDX originated from South American countries that were genetically and biologically characterized and may be used in precision medicine studies. PDX expressing AR and/or GR are powerful tools to evaluate different endocrine treatment combinations even in TN tumors.
Collapse
Affiliation(s)
- Gabriela Pataccini
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Andrés Elia
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Gonzalo Sequeira
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Luisa Ambrosio
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Marcela Coianis
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Caroline A Lamb
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Paola A Rojas
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | | | - Javier Burruchaga
- Hospital de Agudos Magdalena V. de Martínez, General Pacheco, Buenos Aires, Argentina
| | - Eunice Spengler
- Hospital de Agudos Magdalena V. de Martínez, General Pacheco, Buenos Aires, Argentina
| | | | - Martin Abba
- Universidad Nacional de La Plata, La Plata, Argentina
| | - Claudia Lanari
- Laboratory of Hormonal Carcinogenesis, IBYME-CONICET, Ciudad Autónoma de Buenos Aires (CABA), V. Obligado 2490, C1428ADN, Buenos Aires, Argentina.
| |
Collapse
|
9
|
Khairani AF, Harmonia S, Chou Y, Alfarafisa NM, Ramadhanti J. Optimizing Xenograft Models for Breast Cancer: A Comparative Analysis of Cell-Derived and Patient-Derived Implantation Techniques in Pre-Clinical Research. BREAST CANCER (DOVE MEDICAL PRESS) 2025; 17:1-10. [PMID: 39811602 PMCID: PMC11727321 DOI: 10.2147/bctt.s490532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/19/2024] [Indexed: 01/16/2025]
Abstract
Purpose The high mortality rate of breast cancer motivates researchers to search for effective treatments. Due to their ability to simulate human conditions, xenograft models such as CDX (Cell line-Derived Xenografts) and PDX (Patient-Derived Xenografts) have gained popularity in pre-clinical research. The choice of xenograft technique is influenced by the type of tumor employed, particularly in more aggressive tumor models like TNBC with metastases. Subcutaneous or orthotopic implantation may influence tumor engraftment rates and the applicability of the models for drug testing. To optimize xenograft models and support the development of breast cancer drugs, selecting a suitable transplantation technique is essential to attaining the best results. Methods This scoping review used PRISMA-Scr methodology to summarize findings from eleven articles published between 2012 and 2024 on pre-clinical trials related to xenograft models for breast cancer considering PDX began traction after 2010. Using specific criteria, the review included studies from electronic platforms. The inclusion criteria ensured relevant English sources were available in full text, while the exclusion criteria eliminated certain types of articles and inadequately comprehensive studies. Results Subcutaneous and orthotopic implantation are critical methods for xenograft models in cancer research. Subcutaneous implantation is less invasive and more manageable but does not fully mimic the tumor's natural environment. Orthotopic implantation accurately mimic the migration, invasion, and molecular characteristics of the original tumor, although the procedure is more complex and requires specialized techniques. The specific research objectives determine their choice, the need for accurate tumor replication, and the testing convenience. Conclusion Orthotopic implantation is the preferable method for developing PDX and CDX models of breast cancer because it closely mimics the tumor microenvironment and metastatic behavior, yielding clinically relevant results for drug testing. Subcutaneous implantation may result in higher engraftment rates, but it cannot accurately represent the complexity of tumors.
Collapse
Affiliation(s)
- Astrid Feinisa Khairani
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Undergraduate Program, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Graduate School of Master Program in Anti Aging and Aesthetic Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Shella Harmonia
- Undergraduate Program, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Yoan Chou
- Graduate School of Master Program in Anti Aging and Aesthetic Medicine, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Nayla Majeda Alfarafisa
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Undergraduate Program, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| | - Julia Ramadhanti
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
- Undergraduate Program, Faculty of Medicine, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
10
|
Lei JT, Dobrolecki LE, Huang C, Srinivasan RR, Vasaikar SV, Lewis AN, Sallas C, Zhao N, Cao J, Landua JD, Moon CI, Liao Y, Hilsenbeck SG, Osborne CK, Rimawi MF, Ellis MJ, Petrosyan V, Wen B, Li K, Saltzman AB, Jain A, Malovannaya A, Wulf GM, Marangoni E, Li S, Kraushaar DC, Wang T, Damodaran S, Zheng X, Meric-Bernstam F, Echeverria GV, Anurag M, Chen X, Welm BE, Welm AL, Zhang B, Lewis MT. Patient-Derived Xenografts of Triple-Negative Breast Cancer Enable Deconvolution and Prediction of Chemotherapy Responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.09.627518. [PMID: 39713418 PMCID: PMC11661147 DOI: 10.1101/2024.12.09.627518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Combination chemotherapy remains essential for clinical management of triple-negative breast cancer (TNBC). Consequently, responses to multiple single agents cannot be delineated at the single patient level, even though some patients might not require all drugs in the combination. Herein, we conduct multi-omic analyses of orthotopic TNBC patient-derived xenografts (PDXs) treated with single agent carboplatin, docetaxel, or the combination. Combination responses were usually no better than the best single agent, with enhanced response in only ~13% of PDX, and apparent antagonism in a comparable percentage. Single-omic comparisons showed largely non-overlapping results between genes associated with single agent and combination treatments that could be validated in independent patient cohorts. Multi-omic analyses of PDXs identified agent-specific biomarkers/biomarker combinations, nominating high Cytokeratin-5 (KRT5) as a general marker of responsiveness. Notably, integrating proteomic with transcriptomic data improved predictive modeling of pathologic complete response to combination chemotherapy. PDXs refractory to all treatments were enriched for signatures of dysregulated mitochondrial function. Targeting this process indirectly in a PDX with HDAC inhibition plus chemotherapy in vivo overcomes chemoresistance. These results suggest possible resistance mechanisms and therapeutic strategies in TNBC to overcome chemoresistance, and potentially allow optimization of chemotherapeutic regimens.
Collapse
Affiliation(s)
- Jonathan T. Lei
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lacey E. Dobrolecki
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chen Huang
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Current affiliation: Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ramakrishnan R. Srinivasan
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Suhas V. Vasaikar
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Current affiliation: Translational Oncology Bioinformatics, Pfizer, Bothell, WA 98021, USA
| | - Alaina N. Lewis
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Christina Sallas
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Na Zhao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jin Cao
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Current affiliation: The MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - John D. Landua
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chang In Moon
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yuxing Liao
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Susan G. Hilsenbeck
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - C. Kent Osborne
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mothaffar F. Rimawi
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Matthew J. Ellis
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Current affiliation: Guardant Health, Palo Alto, CA 94304, USA
| | - Varduhi Petrosyan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bo Wen
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Current affiliation: Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Kai Li
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Current affiliation: Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alexander B. Saltzman
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX 77030, USA
| | - Antrix Jain
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX 77030, USA
| | - Anna Malovannaya
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Gerburg M. Wulf
- Cancer Research Institute, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Elisabetta Marangoni
- Laboratory of Preclinical investigation, Translational Research Department, Institut Curie, PSL University, 26 Rue d’Ulm, Paris 75005, France
| | - Shunqiang Li
- Siteman Cancer Center, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Daniel C. Kraushaar
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tao Wang
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | - Gloria V. Echeverria
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Radiation Oncology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Meenakshi Anurag
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xi Chen
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bryan E. Welm
- Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Alana L. Welm
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Bing Zhang
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michael T. Lewis
- Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Radiology, Baylor College of Medicine, Houston, TX 77030, USA
- Lead contact
| |
Collapse
|
11
|
Marangoni E. Patient-Derived Xenografts of Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:109-121. [PMID: 39821023 DOI: 10.1007/978-3-031-70875-6_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Patient-derived xenografts (PDX) of breast cancer, obtained from the engraftment of tumour samples into immunodeficient mice, are the most effective preclinical models for studying the biology of human breast cancer and for the evaluation of new anti-cancer treatments. Notably, breast cancer PDX preserve the phenotypic and molecular characteristics of the donor tumours and reproduce the diversity of breast cancer. This preservation of breast cancer biology involves a number of different aspects, including tumour architecture and morphology, patterns of genomic alterations and gene expression, mutational status, and intra-tumour heterogeneity. For these reasons, these models have a strong predictive value in the translation of cancer therapeutics into clinical settings and can be considered as powerful and clinically relevant research tools for the identification of new treatments, mechanisms of drug resistance, and predictive biomarkers. PDX models have also been successfully used to analyse breast cancer metastasis and persister cancer cells surviving chemotherapy. Limitations of breast cancer PDX include the lack of a human immune system and the low take rate, especially for estrogen receptor (ER) and HER2-positive subtypes.
Collapse
Affiliation(s)
- Elisabetta Marangoni
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, Paris, France.
| |
Collapse
|
12
|
Manore S, Zhuang C, Najjar MK, Wong GL, Bindal S, Watabe K, Lin J, Lo HW. Co-Inhibition of tGLI1 and GP130 Using FDA-Approved Ketoconazole and Bazedoxifene Is Synergistic Against the Growth and Metastasis of HER2-Enriched and Triple-Negative Breast Cancers. Cells 2024; 13:2087. [PMID: 39768178 PMCID: PMC11674475 DOI: 10.3390/cells13242087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Breast cancer stem cells (CSCs) are resistant to most cancer therapeutics and contribute to tumor recurrence and metastasis. Two breast CSC-promoting transcription factors, truncated glioma-associated oncogene homolog 1 (tGLI1) and signal transducer and activator of transcription 3 (STAT3), have been reported to be frequently co-expressed in HER2-enriched breast cancer and triple-negative breast cancer (TNBC), undergo protein-protein interactions for gene regulation and activation, and functionally cooperate to promote breast CSCs. STAT3 can be activated by activated interleukin-6 receptor/glycoprotein-130 (IL-6R/GP130). Co-targeting of tGLI1 and IL-6R/GP130 has not been investigated in breast cancer or any tumor type. Here, we report that tGLI1 and GP130 are co-overexpressed in the majority of HER2-enriched breast cancers and TNBCs at 53.8% and 44.4%, respectively. tGLI1+IL-6/IL-6R/GP130 signaling is frequently co-enriched and co-activated in HER2-enriched breast cancer and TNBC when compared to luminal subtypes. tGLI1+GP130 co-overexpression strongly promotes CSCs of HER2-enriched breast cancer and TNBC. FDA-approved tGLI1 inhibitor Ketoconazole and GP130 inhibitor Bazedoxifene synergize against breast cancer proliferation and CSC phenotypes in vitro and reduce TNBC tumor growth and metastatic burden in vivo. Our study demonstrates, for the first time, that co-targeting tGLI1 and IL-6R/GP130/STAT3 signaling pathways is synergistic against HER2-enriched breast cancer and TNBC, warranting future clinical investigations.
Collapse
Affiliation(s)
- Sara Manore
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (S.M.); (C.Z.); (M.K.N.); (G.L.W.); (S.B.)
- Wake Forest Graduate School of Biomedical Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Chuling Zhuang
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (S.M.); (C.Z.); (M.K.N.); (G.L.W.); (S.B.)
- Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Mariana K. Najjar
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (S.M.); (C.Z.); (M.K.N.); (G.L.W.); (S.B.)
- Wake Forest Graduate School of Biomedical Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
- Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Grace L. Wong
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (S.M.); (C.Z.); (M.K.N.); (G.L.W.); (S.B.)
- Wake Forest Graduate School of Biomedical Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Shivani Bindal
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (S.M.); (C.Z.); (M.K.N.); (G.L.W.); (S.B.)
| | - Kounosuke Watabe
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA;
| | - Jiayuh Lin
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Hui-Wen Lo
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (S.M.); (C.Z.); (M.K.N.); (G.L.W.); (S.B.)
- Wake Forest Graduate School of Biomedical Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
- Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA;
| |
Collapse
|
13
|
Xu L, Peng F, Luo Q, Ding Y, Yuan F, Zheng L, He W, Zhang SS, Fu X, Liu J, Mutlu AS, Wang S, Nehring RB, Li X, Tang Q, Li C, Lv X, Dobrolecki LE, Zhang W, Han D, Zhao N, Jaehnig E, Wang J, Wu W, Graham DA, Li Y, Chen R, Peng W, Chen Y, Catic A, Zhang Z, Zhang B, Mustoe AM, Koong AC, Miles G, Lewis MT, Wang MC, Rosenberg SM, O'Malley BW, Westbrook TF, Xu H, Zhang XHF, Osborne CK, Li JB, Ellis MJ, Rimawi MF, Rosen JM, Chen X. IRE1α silences dsRNA to prevent taxane-induced pyroptosis in triple-negative breast cancer. Cell 2024; 187:7248-7266.e34. [PMID: 39419025 PMCID: PMC11645245 DOI: 10.1016/j.cell.2024.09.032] [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: 12/06/2022] [Revised: 07/10/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024]
Abstract
Chemotherapy is often combined with immune checkpoint inhibitor (ICIs) to enhance immunotherapy responses. Despite the approval of chemo-immunotherapy in multiple human cancers, many immunologically cold tumors remain unresponsive. The mechanisms determining the immunogenicity of chemotherapy are elusive. Here, we identify the ER stress sensor IRE1α as a critical checkpoint that restricts the immunostimulatory effects of taxane chemotherapy and prevents the innate immune recognition of immunologically cold triple-negative breast cancer (TNBC). IRE1α RNase silences taxane-induced double-stranded RNA (dsRNA) through regulated IRE1-dependent decay (RIDD) to prevent NLRP3 inflammasome-dependent pyroptosis. Inhibition of IRE1α in Trp53-/- TNBC allows taxane to induce extensive dsRNAs that are sensed by ZBP1, which in turn activates NLRP3-GSDMD-mediated pyroptosis. Consequently, IRE1α RNase inhibitor plus taxane converts PD-L1-negative, ICI-unresponsive TNBC tumors into PD-L1high immunogenic tumors that are hyper-sensitive to ICI. We reveal IRE1α as a cancer cell defense mechanism that prevents taxane-induced danger signal accumulation and pyroptotic cell death.
Collapse
Affiliation(s)
- Longyong Xu
- Department of Experimental Therapeutics, James P. Allison Institute, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Fanglue Peng
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Qin Luo
- Department of Experimental Therapeutics, James P. Allison Institute, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Yao Ding
- Department of Experimental Therapeutics, James P. Allison Institute, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Fei Yuan
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Liting Zheng
- Therapeutic Innovation Center (THINC), and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Wei He
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sophie S Zhang
- Department of Chemistry, Rice University, Houston, TX 77005, USA
| | - Xin Fu
- Department of Pathology, Xijing Hospital, Xi'an, Shaanxi 710032, China
| | - Jin Liu
- Department of Pathology, Xijing Hospital, Xi'an, Shaanxi 710032, China
| | - Ayse Sena Mutlu
- Therapeutic Innovation Center (THINC), and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shuyue Wang
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ralf Bernd Nehring
- Therapeutic Innovation Center (THINC), and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xingyu Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Qianzi Tang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Catherine Li
- Department of Experimental Therapeutics, James P. Allison Institute, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiangdong Lv
- Department of Experimental Therapeutics, James P. Allison Institute, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Lacey E Dobrolecki
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Weijie Zhang
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dong Han
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Na Zhao
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Eric Jaehnig
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jingyi Wang
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Weiche Wu
- Department of Experimental Therapeutics, James P. Allison Institute, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Davis A Graham
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yumei Li
- Therapeutic Innovation Center (THINC), and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Rui Chen
- Therapeutic Innovation Center (THINC), and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Weiyi Peng
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| | - Yiwen Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Andre Catic
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhibin Zhang
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bing Zhang
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Anthony M Mustoe
- Therapeutic Innovation Center (THINC), and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Albert C Koong
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - George Miles
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michael T Lewis
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Meng C Wang
- HHMI Janelia Research Campus, Ashburn, VA 20147, USA
| | - Susan M Rosenberg
- Therapeutic Innovation Center (THINC), and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Thomas F Westbrook
- Therapeutic Innovation Center (THINC), and Marrs McLean Department of Biochemistry and Molecular Pharmacology, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Han Xu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiang H-F Zhang
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - C Kent Osborne
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jin Billy Li
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Matthew J Ellis
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mothaffar F Rimawi
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jeffrey M Rosen
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xi Chen
- Department of Experimental Therapeutics, James P. Allison Institute, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dun L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
14
|
Rinkenbaugh AL, Qi Y, Cai S, Shao J, Hancock F, Jeter-Jones S, Zhang X, Powell E, Huo L, Lau R, Fu C, Gould R, den Hollander P, Ravenberg EE, White JB, Rauch GM, Arun B, Yam C, Thompson AM, Echeverria GV, Moulder SL, Symmans WF, Chang JT, Piwnica-Worms H. An annotated biobank of triple negative breast cancer patient-derived xenografts featuring treatment-naïve and longitudinal samples throughout neoadjuvant chemotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.25.625287. [PMID: 39651299 PMCID: PMC11623626 DOI: 10.1101/2024.11.25.625287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Triple negative breast cancer (TNBC) that fails to respond to neoadjuvant chemotherapy (NACT) can be lethal. Developing effective strategies to eradicate chemoresistant disease requires experimental models that recapitulate the heterogeneity characteristic of TNBC. To that end, we established a biobank of 92 orthotopic patient-derived xenograft (PDX) models of TNBC from the tumors of 75 patients enrolled in the ARTEMIS clinical trial ( NCT02276443 ) at MD Anderson Cancer Center, including 12 longitudinal sets generated from serial patient biopsies collected throughout NACT and from metastatic disease. Models were established from both chemosensitive and chemoresistant tumors, and nearly 30% of PDX models were capable of lung metastasis. Comprehensive molecular profiling demonstrated conservation of genomes and transcriptomes between patient and corresponding PDX tumors, with representation of all major transcriptional subtypes. Transcriptional changes observed in the longitudinal PDX models highlight dysregulation in pathways associated with DNA integrity, extracellular matrix interactions, the ubiquitin-proteasome system, epigenetics, and inflammatory signaling. These alterations reveal a complex network of adaptations associated with chemoresistance. This PDX biobank provides a valuable resource for tackling the most pressing issues facing the clinical management of TNBC, namely chemoresistance and metastasis.
Collapse
|
15
|
Fernando W, Cruickshank BM, Arun RP, MacLean MR, Cahill HF, Morales-Quintanilla F, Dean CA, Wasson MCD, Dahn ML, Coyle KM, Walker OL, Power Coombs MR, Marcato P. ALDH1A3 is the switch that determines the balance of ALDH + and CD24 -CD44 + cancer stem cells, EMT-MET, and glucose metabolism in breast cancer. Oncogene 2024; 43:3151-3169. [PMID: 39251846 PMCID: PMC11493680 DOI: 10.1038/s41388-024-03156-4] [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: 05/02/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024]
Abstract
Plasticity is an inherent feature of cancer stem cells (CSCs) and regulates the balance of key processes required at different stages of breast cancer progression, including epithelial-to-mesenchymal transition (EMT) versus mesenchymal-to-epithelial transition (MET), and glycolysis versus oxidative phosphorylation. Understanding the key factors that regulate the switch between these processes could lead to novel therapeutic strategies that limit tumor progression. We found that aldehyde dehydrogenase 1A3 (ALDH1A3) regulates these cancer-promoting processes and the abundance of the two distinct breast CSC populations defined by high ALDH activity and CD24-CD44+ cell surface expression. While ALDH1A3 increases ALDH+ breast cancer cells, it inversely suppresses the CD24-CD44+ population by retinoic acid signaling-mediated gene expression changes. This switch in CSC populations induced by ALDH1A3 was paired with decreased migration but increased invasion and an intermediate EMT phenotype. We also demonstrate that ALDH1A3 increases oxidative phosphorylation and decreases glycolysis and reactive oxygen species (ROS). The effects of ALDH1A3 reduction were countered with the glycolysis inhibitor 2-deoxy-D-glucose (2DG). In cell culture and tumor xenograft models, 2DG suppresses the increase in the CD24-CD44+ population and ROS induced by ALDH1A3 knockdown. Combined inhibition of ALDH1A3 and glycolysis best reduces breast tumor growth and tumor-initiating cells, suggesting that the combination of targeting ALDH1A3 and glycolysis has therapeutic potential for limiting CSCs and tumor progression. Together, these findings identify ALDH1A3 as a key regulator of processes required for breast cancer progression and depletion of ALDH1A3 makes breast cancer cells more susceptible to glycolysis inhibition.
Collapse
Affiliation(s)
- Wasundara Fernando
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- Department of Biology, Acadia University, Wolfville, NS, Canada
| | - Brianne M Cruickshank
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- Department of Surgery, Dalhousie University, Halifax, NS, Canada
| | - Raj Pranap Arun
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Maya R MacLean
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Hannah F Cahill
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | | | - Cheryl A Dean
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | | | - Margaret L Dahn
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Krysta M Coyle
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- Department of Molecular Biology & Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Olivia L Walker
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Melanie R Power Coombs
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- Department of Biology, Acadia University, Wolfville, NS, Canada
| | - Paola Marcato
- Department of Pathology, Dalhousie University, Halifax, NS, Canada.
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada.
- Nova Scotia Health Authority, Halifax, NS, Canada.
| |
Collapse
|
16
|
Winkler J, Tan W, Diadhiou CM, McGinnis CS, Abbasi A, Hasnain S, Durney S, Atamaniuc E, Superville D, Awni L, Lee JV, Hinrichs JH, Wagner PS, Singh N, Hein MY, Borja M, Detweiler AM, Liu SY, Nanjaraj A, Sitarama V, Rugo HS, Neff N, Gartner ZJ, Oliveira Pisco A, Goga A, Darmanis S, Werb Z. Single-cell analysis of breast cancer metastasis reveals epithelial-mesenchymal plasticity signatures associated with poor outcomes. J Clin Invest 2024; 134:e164227. [PMID: 39225101 PMCID: PMC11364385 DOI: 10.1172/jci164227] [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: 08/04/2022] [Accepted: 06/11/2024] [Indexed: 09/04/2024] Open
Abstract
Metastasis is the leading cause of cancer-related deaths. It is unclear how intratumor heterogeneity (ITH) contributes to metastasis and how metastatic cells adapt to distant tissue environments. The study of these adaptations is challenged by the limited access to patient material and a lack of experimental models that appropriately recapitulate ITH. To investigate metastatic cell adaptations and the contribution of ITH to metastasis, we analyzed single-cell transcriptomes of matched primary tumors and metastases from patient-derived xenograft models of breast cancer. We found profound transcriptional differences between the primary tumor and metastatic cells. Primary tumors upregulated several metabolic genes, whereas motility pathway genes were upregulated in micrometastases, and stress response signaling was upregulated during progression. Additionally, we identified primary tumor gene signatures that were associated with increased metastatic potential and correlated with patient outcomes. Immune-regulatory control pathways were enriched in poorly metastatic primary tumors, whereas genes involved in epithelial-mesenchymal transition were upregulated in highly metastatic tumors. We found that ITH was dominated by epithelial-mesenchymal plasticity (EMP), which presented as a dynamic continuum with intermediate EMP cell states characterized by specific genes such as CRYAB and S100A2. Elevated expression of an intermediate EMP signature correlated with worse patient outcomes. Our findings identified inhibition of the intermediate EMP cell state as a potential therapeutic target to block metastasis.
Collapse
Affiliation(s)
- Juliane Winkler
- Department of Anatomy and
- Department of Cell and Tissue Biology, UCSF, San Francisco, California, USA
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Weilun Tan
- Chan Zuckerberg Biohub SF, San Francisco, California, USA
| | | | | | | | | | - Sophia Durney
- Department of Cell and Tissue Biology, UCSF, San Francisco, California, USA
| | - Elena Atamaniuc
- Department of Cell and Tissue Biology, UCSF, San Francisco, California, USA
| | - Daphne Superville
- Department of Cell and Tissue Biology, UCSF, San Francisco, California, USA
| | | | - Joyce V. Lee
- Department of Cell and Tissue Biology, UCSF, San Francisco, California, USA
| | - Johanna H. Hinrichs
- Department of Anatomy and
- Institute of Internal Medicine D, Medical Cell Biology, University Hospital Münster, Münster, Germany
| | - Patrick S. Wagner
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Namrata Singh
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Marco Y. Hein
- Chan Zuckerberg Biohub SF, San Francisco, California, USA
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Medical University of Vienna, Max Perutz Labs, Vienna, Austria
| | - Michael Borja
- Chan Zuckerberg Biohub SF, San Francisco, California, USA
| | | | | | | | | | - Hope S. Rugo
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
| | - Norma Neff
- Chan Zuckerberg Biohub SF, San Francisco, California, USA
| | - Zev J. Gartner
- Department of Pharmaceutical Chemistry, UCSF, San Francisco, California, USA
- Chan Zuckerberg Biohub Investigator, San Francisco, California, USA
| | | | - Andrei Goga
- Department of Cell and Tissue Biology, UCSF, San Francisco, California, USA
- Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, California, USA
| | - Spyros Darmanis
- Chan Zuckerberg Biohub SF, San Francisco, California, USA
- Genentech, South San Francisco, California, USA
| | | |
Collapse
|
17
|
Pedroza DA, Gao Y, Zhang XHF, Rosen JM. Leveraging preclinical models of metastatic breast cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189163. [PMID: 39084494 PMCID: PMC11390310 DOI: 10.1016/j.bbcan.2024.189163] [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: 11/17/2023] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024]
Abstract
Women that present to the clinic with established breast cancer metastases have limited treatment options. Yet, the majority of preclinical studies are actually not directed at developing treatment regimens for established metastatic disease. In this review we will discuss the current state of preclinical macro-metastatic breast cancer models, including, but not limited to syngeneic GEMM, PDX and xenografts. Challenges within these models which are often overlooked include fluorophore-immunogenic neoantigens, differences in experimental vs spontaneous metastasis and tumor heterogeneity. Furthermore, due to cell plasticity in the tumor immune microenvironment (TIME) of the metastatic landscape, the treatment efficacy of newly approved immune checkpoint blockade (ICB) may differ in metastatic sites as compared to primary localized tumors.
Collapse
Affiliation(s)
- Diego A Pedroza
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, United States of America; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States of America
| | - Yang Gao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, United States of America; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States of America
| | - Xiang H-F Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, United States of America; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States of America
| | - Jeffrey M Rosen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States of America.
| |
Collapse
|
18
|
Tosca EM, Ronchi D, Rocchetti M, Magni P. Predicting Tumor Volume Doubling Time and Progression-Free Survival in Untreated Patients from Patient-Derived-Xenograft (PDX) Models: A Translational Model-Based Approach. AAPS J 2024; 26:92. [PMID: 39117850 DOI: 10.1208/s12248-024-00960-4] [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] [Received: 03/13/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Tumor volume doubling time (TVDT) has been shown to be a potential surrogate marker of biological tumor activity. However, its availability in clinics is strongly limited due to ethical and practical reasons, as its assessment requires at least two subsequent tumor volume measurements in untreated patients. Here, a translational modeling framework to predict TVDT distributions in untreated cancer patient populations from tumor growth data in patient-derived xenograft (PDX) mice is proposed. Eleven solid cancer types were considered. For each of them, a set of tumor growth studies in PDX mice was selected and analyzed through a mathematical model to characterize the distribution of the exponential tumor growth rate in mice. Then, assuming an exponential growth of the tumor mass in humans, the growth rates were scaled from PDX mice to humans through an allometric scaling approach and used to predict TVDTs in untreated patients. A very good agreement was found between model predicted and clinically observed TVDTs, with 91% of the predicted TVDT medians fell within 1.5-fold of observations. Further, exploiting the intrinsic relationship between tumor growth dynamics and progression free survival (PFS), the exponential growth rates in humans were used to generate the expected PFS curves in absence of anticancer treatment. Predicted curves were extremely close to published PFS data from studies involving patient cohorts treated with supportive care or low effective therapies. The proposed approach shows promise as a potential tool to increase knowledge about TVDT in humans without the need of directly measuring tumor dimensions in untreated patients, and to predict PFS curves in untreated patients, that could fill the absence of placebo-controlled arms against which to compare treaded arms during clinical trials. However, further validation and refinement are needed to fully assess its effectiveness in this regard.
Collapse
Affiliation(s)
- E M Tosca
- Dipartimento Di Ingegneria Industriale E Dell'Informazione, Università Degli Studi Di Pavia, 27100, Pavia, Italy
| | - D Ronchi
- Dipartimento Di Ingegneria Industriale E Dell'Informazione, Università Degli Studi Di Pavia, 27100, Pavia, Italy
| | | | - P Magni
- Dipartimento Di Ingegneria Industriale E Dell'Informazione, Università Degli Studi Di Pavia, 27100, Pavia, Italy.
| |
Collapse
|
19
|
Khorsandi D, Yang JW, Foster S, Khosravi S, Hoseinzadeh N, Zarei F, Lee YB, Runa F, Gangrade A, Voskanian L, Adnan D, Zhu Y, Wang Z, Jucaud V, Dokmeci MR, Shen X, Bishehsari F, Kelber JA, Khademhosseini A, de Barros NR. Patient-Derived Organoids as Therapy Screening Platforms in Cancer Patients. Adv Healthc Mater 2024; 13:e2302331. [PMID: 38359321 PMCID: PMC11324859 DOI: 10.1002/adhm.202302331] [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: 07/21/2023] [Revised: 11/28/2023] [Indexed: 02/17/2024]
Abstract
Patient-derived organoids (PDOs) developed ex vivo and in vitro are increasingly used for therapeutic screening. They provide a more physiologically relevant model for drug discovery and development compared to traditional cell lines. However, several challenges remain to be addressed to fully realize the potential of PDOs in therapeutic screening. This paper summarizes recent advancements in PDO development and the enhancement of PDO culture models. This is achieved by leveraging materials engineering and microfabrication technologies, including organs-on-a-chip and droplet microfluidics. Additionally, this work discusses the application of PDOs in therapy screening to meet diverse requirements and overcome bottlenecks in cancer treatment. Furthermore, this work introduces tools for data processing and analysis of organoids, along with their microenvironment. These tools aim to achieve enhanced readouts. Finally, this work explores the challenges and future perspectives of using PDOs in drug development and personalized screening for cancer patients.
Collapse
Affiliation(s)
- Danial Khorsandi
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Jia-Wei Yang
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Samuel Foster
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Safoora Khosravi
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Negar Hoseinzadeh
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Fahimeh Zarei
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Yun Bin Lee
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Farhana Runa
- California State University Northridge, 18111 Nordhoff Street, Northridge, California, USA
| | - Ankit Gangrade
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Leon Voskanian
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Darbaz Adnan
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush Medical College, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Zhaohui Wang
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710 USA
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Mehmet Remzi Dokmeci
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Xiling Shen
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Faraz Bishehsari
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush Medical College, Rush University Medical Center, Chicago, IL, 60612, USA
- Division of Digestive Diseases, Rush Center for Integrated Microbiome & Chronobiology Research, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Jonathan A. Kelber
- California State University Northridge, 18111 Nordhoff Street, Northridge, California, USA
- Baylor University, 101 Bagby Ave, Waco, Texas, USA
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Natan Roberto de Barros
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| |
Collapse
|
20
|
Cheung A, Chenoweth AM, Johansson A, Laddach R, Guppy N, Trendell J, Esapa B, Mavousian A, Navarro-Llinas B, Haider S, Romero-Clavijo P, Hoffmann RM, Andriollo P, Rahman KM, Jackson P, Tsoka S, Irshad S, Roxanis I, Grigoriadis A, Thurston DE, Lord CJ, Tutt ANJ, Karagiannis SN. Anti-EGFR Antibody-Drug Conjugate Carrying an Inhibitor Targeting CDK Restricts Triple-Negative Breast Cancer Growth. Clin Cancer Res 2024; 30:3298-3315. [PMID: 38772416 PMCID: PMC11292198 DOI: 10.1158/1078-0432.ccr-23-3110] [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: 10/12/2023] [Revised: 03/06/2024] [Accepted: 05/15/2024] [Indexed: 05/23/2024]
Abstract
PURPOSE Anti-EGFR antibodies show limited response in breast cancer, partly due to activation of compensatory pathways. Furthermore, despite the clinical success of cyclin-dependent kinase (CDK) 4/6 inhibitors in hormone receptor-positive tumors, aggressive triple-negative breast cancers (TNBC) are largely resistant due to CDK2/cyclin E expression, whereas free CDK2 inhibitors display normal tissue toxicity, limiting their therapeutic application. A cetuximab-based antibody drug conjugate (ADC) carrying a CDK inhibitor selected based on oncogene dysregulation, alongside patient subgroup stratification, may provide EGFR-targeted delivery. EXPERIMENTAL DESIGN Expressions of G1/S-phase cell cycle regulators were evaluated alongside EGFR in breast cancer. We conjugated cetuximab with CDK inhibitor SNS-032, for specific delivery to EGFR-expressing cells. We assessed ADC internalization and its antitumor functions in vitro and in orthotopically grown basal-like/TNBC xenografts. RESULTS Transcriptomic (6,173 primary, 27 baseline, and matched post-chemotherapy residual tumors), single-cell RNA sequencing (150,290 cells, 27 treatment-naïve tumors), and spatial transcriptomic (43 tumor sections, 22 TNBCs) analyses confirmed expression of CDK2 and its cyclin partners in basal-like/TNBCs, associated with EGFR. Spatiotemporal live-cell imaging and super-resolution confocal microscopy demonstrated ADC colocalization with late lysosomal clusters. The ADC inhibited cell cycle progression, induced cytotoxicity against high EGFR-expressing tumor cells, and bystander killing of neighboring EGFR-low tumor cells, but minimal effects on immune cells. Despite carrying a small molar fraction (1.65%) of the SNS-032 inhibitor, the ADC restricted EGFR-expressing spheroid and cell line/patient-derived xenograft tumor growth. CONCLUSIONS Exploiting EGFR overexpression, and dysregulated cell cycle in aggressive and treatment-refractory tumors, a cetuximab-CDK inhibitor ADC may provide selective and efficacious delivery of cell cycle-targeted agents to basal-like/TNBCs, including chemotherapy-resistant residual disease.
Collapse
Affiliation(s)
- Anthony Cheung
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, London, United Kingdom
- St. John’s Institute of Dermatology, School of Basic and Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Alicia M. Chenoweth
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, London, United Kingdom
- St. John’s Institute of Dermatology, School of Basic and Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Annelie Johansson
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, London, United Kingdom
- Cancer Bioinformatics, School of Cancer and Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, London, United Kingdom
| | - Roman Laddach
- St. John’s Institute of Dermatology, School of Basic and Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, Guy’s Hospital, London, United Kingdom
- Department of Informatics, Faculty of Natural, Mathematical and Engineering Sciences, King’s College London, London, United Kingdom
| | - Naomi Guppy
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Jennifer Trendell
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, London, United Kingdom
| | - Benjamina Esapa
- St. John’s Institute of Dermatology, School of Basic and Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Antranik Mavousian
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Blanca Navarro-Llinas
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, London, United Kingdom
| | - Syed Haider
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Pablo Romero-Clavijo
- St. John’s Institute of Dermatology, School of Basic and Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Ricarda M. Hoffmann
- St. John’s Institute of Dermatology, School of Basic and Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, Guy’s Hospital, London, United Kingdom
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Paolo Andriollo
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, London, United Kingdom
| | - Khondaker M. Rahman
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, London, United Kingdom
| | - Paul Jackson
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, London, United Kingdom
| | - Sophia Tsoka
- Department of Informatics, Faculty of Natural, Mathematical and Engineering Sciences, King’s College London, London, United Kingdom
| | - Sheeba Irshad
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, London, United Kingdom
| | - Ioannis Roxanis
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Anita Grigoriadis
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, London, United Kingdom
- Cancer Bioinformatics, School of Cancer and Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, London, United Kingdom
| | - David E. Thurston
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, King’s College London, London, United Kingdom
| | - Christopher J. Lord
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Andrew N. J. Tutt
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, London, United Kingdom
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Sophia N. Karagiannis
- Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, London, United Kingdom
- St. John’s Institute of Dermatology, School of Basic and Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, Guy’s Hospital, London, United Kingdom
| |
Collapse
|
21
|
White BS, Woo XY, Koc S, Sheridan T, Neuhauser SB, Wang S, Evrard YA, Chen L, Foroughi pour A, Landua JD, Mashl RJ, Davies SR, Fang B, Raso MG, Evans KW, Bailey MH, Chen Y, Xiao M, Rubinstein JC, Sanderson BJ, Lloyd MW, Domanskyi S, Dobrolecki LE, Fujita M, Fujimoto J, Xiao G, Fields RC, Mudd JL, Xu X, Hollingshead MG, Jiwani S, Acevedo S, Davis-Dusenbery BN, Robinson PN, Moscow JA, Doroshow JH, Mitsiades N, Kaochar S, Pan CX, Carvajal-Carmona LG, Welm AL, Welm BE, Govindan R, Li S, Davies MA, Roth JA, Meric-Bernstam F, Xie Y, Herlyn M, Ding L, Lewis MT, Bult CJ, Dean DA, Chuang JH. A Pan-Cancer Patient-Derived Xenograft Histology Image Repository with Genomic and Pathologic Annotations Enables Deep Learning Analysis. Cancer Res 2024; 84:2060-2072. [PMID: 39082680 PMCID: PMC11217732 DOI: 10.1158/0008-5472.can-23-1349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 10/13/2023] [Accepted: 03/27/2024] [Indexed: 08/04/2024]
Abstract
Patient-derived xenografts (PDX) model human intra- and intertumoral heterogeneity in the context of the intact tissue of immunocompromised mice. Histologic imaging via hematoxylin and eosin (H&E) staining is routinely performed on PDX samples, which could be harnessed for computational analysis. Prior studies of large clinical H&E image repositories have shown that deep learning analysis can identify intercellular and morphologic signals correlated with disease phenotype and therapeutic response. In this study, we developed an extensive, pan-cancer repository of >1,000 PDX and paired parental tumor H&E images. These images, curated from the PDX Development and Trial Centers Research Network Consortium, had a range of associated genomic and transcriptomic data, clinical metadata, pathologic assessments of cell composition, and, in several cases, detailed pathologic annotations of neoplastic, stromal, and necrotic regions. The amenability of these images to deep learning was highlighted through three applications: (i) development of a classifier for neoplastic, stromal, and necrotic regions; (ii) development of a predictor of xenograft-transplant lymphoproliferative disorder; and (iii) application of a published predictor of microsatellite instability. Together, this PDX Development and Trial Centers Research Network image repository provides a valuable resource for controlled digital pathology analysis, both for the evaluation of technical issues and for the development of computational image-based methods that make clinical predictions based on PDX treatment studies. Significance: A pan-cancer repository of >1,000 patient-derived xenograft hematoxylin and eosin-stained images will facilitate cancer biology investigations through histopathologic analysis and contributes important model system data that expand existing human histology repositories.
Collapse
Affiliation(s)
- Brian S. White
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut.
| | - Xing Yi Woo
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut.
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
| | - Soner Koc
- Velsera, Charlestown, Massachusetts.
| | - Todd Sheridan
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut.
| | | | - Shidan Wang
- University of Texas Southwestern Medical Center, Dallas, Texas.
| | - Yvonne A. Evrard
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland.
| | - Li Chen
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland.
| | - Ali Foroughi pour
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut.
| | | | - R. Jay Mashl
- Washington University School of Medicine, St. Louis, Missouri.
| | | | - Bingliang Fang
- University of Texas MD Anderson Cancer Center, Houston, Texas.
| | | | - Kurt W. Evans
- University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Matthew H. Bailey
- Simmons Center for Cancer Research, Brigham Young University, Provo, Utah.
| | - Yeqing Chen
- The Wistar Institute, Philadelphia, Pennsylvania.
| | - Min Xiao
- The Wistar Institute, Philadelphia, Pennsylvania.
| | | | | | | | - Sergii Domanskyi
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut.
| | | | - Maihi Fujita
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah.
| | - Junya Fujimoto
- University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Guanghua Xiao
- University of Texas Southwestern Medical Center, Dallas, Texas.
| | - Ryan C. Fields
- Washington University School of Medicine, St. Louis, Missouri.
| | | | - Xiaowei Xu
- The Wistar Institute, Philadelphia, Pennsylvania.
| | | | - Shahanawaz Jiwani
- Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland.
| | | | | | | | - Peter N. Robinson
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut.
| | | | | | | | | | | | | | - Alana L. Welm
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah.
| | - Bryan E. Welm
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah.
| | | | - Shunqiang Li
- Washington University School of Medicine, St. Louis, Missouri.
| | | | - Jack A. Roth
- University of Texas MD Anderson Cancer Center, Houston, Texas.
| | | | - Yang Xie
- University of Texas Southwestern Medical Center, Dallas, Texas.
| | | | - Li Ding
- Washington University School of Medicine, St. Louis, Missouri.
| | | | | | | | - Jeffrey H. Chuang
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut.
| |
Collapse
|
22
|
Cocco E, de Stanchina E. Patient-Derived-Xenografts in Mice: A Preclinical Platform for Cancer Research. Cold Spring Harb Perspect Med 2024; 14:a041381. [PMID: 37696659 PMCID: PMC11216185 DOI: 10.1101/cshperspect.a041381] [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] [Indexed: 09/13/2023]
Abstract
The use of patient-derived xenografts (PDXs) has dramatically improved drug development programs. PDXs (1) reproduce the pathological features and the genomic profile of the parental tumors more precisely than other preclinical models, and (2) more faithfully predict therapy response. However, PDXs have limitations. These include the inability to completely capture tumor heterogeneity and the role of the immune system, the low engraftment efficiency of certain tumor types, and the consequences of the human-host interactions. Recently, the use of novel mouse strains and specialized engraftment techniques has enabled the generation of "humanized" PDXs, partially overcoming such limitations. Importantly, establishing, characterizing, and maintaining PDXs is costly and requires a significant regulatory, administrative, clinical, and laboratory infrastructure. In this review, we will retrace the historical milestones that led to the implementation of PDXs for cancer research, review the most recent innovations in the field, and discuss future avenues to tackle deficiencies that still exist.
Collapse
Affiliation(s)
- Emiliano Cocco
- University of Miami, Miller School of Medicine, Department of Biochemistry and Molecular Biology, Sylvester Comprehensive Cancer Center, Miami, Florida 33136, USA
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| |
Collapse
|
23
|
Wang R, Khatpe AS, Kumar B, Mang HE, Batic K, Adebayo AK, Nakshatri H. Mutant RAS-driven Secretome Causes Skeletal Muscle Defects in Breast Cancer. CANCER RESEARCH COMMUNICATIONS 2024; 4:1282-1295. [PMID: 38651826 PMCID: PMC11094532 DOI: 10.1158/2767-9764.crc-24-0045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/28/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
Cancer-induced skeletal muscle defects differ in severity between individuals with the same cancer type. Cancer subtype-specific genomic aberrations are suggested to mediate these differences, but experimental validation studies are very limited. We utilized three different breast cancer patient-derived xenograft (PDX) models to correlate cancer subtype with skeletal muscle defects. PDXs were derived from brain metastasis of triple-negative breast cancer (TNBC), estrogen receptor-positive/progesterone receptor-positive (ER+/PR+) primary breast cancer from a BRCA2-mutation carrier, and pleural effusion from an ER+/PR- breast cancer. While impaired skeletal muscle function as measured through rotarod performance and reduced levels of circulating and/or skeletal muscle miR-486 were common across all three PDXs, only TNBC-derived PDX activated phospho-p38 in skeletal muscle. To further extend these results, we generated transformed variants of human primary breast epithelial cells from healthy donors using HRASG12V or PIK3CAH1047R mutant oncogenes. Mutations in RAS oncogene or its modulators are found in approximately 37% of metastatic breast cancers, which is often associated with skeletal muscle defects. Although cells transformed with both oncogenes generated adenocarcinomas in NSG mice, only HRASG12V-derived tumors caused skeletal muscle defects affecting rotarod performance, skeletal muscle contraction force, and miR-486, Pax7, pAKT, and p53 levels in skeletal muscle. Circulating levels of the chemokine CXCL1 were elevated only in animals with tumors containing HRASG12V mutation. Because RAS pathway aberrations are found in 19% of cancers, evaluating skeletal muscle defects in the context of genomic aberrations in cancers, particularly RAS pathway mutations, may accelerate development of therapeutic modalities to overcome cancer-induced systemic effects. SIGNIFICANCE Mutant RAS- and PIK3CA-driven breast cancers distinctly affect the function of skeletal muscle. Therefore, research and therapeutic targeting of cancer-induced systemic effects need to take aberrant cancer genome into consideration.
Collapse
Affiliation(s)
- Ruizhong Wang
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Aditi S. Khatpe
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Brijesh Kumar
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Henry Elmer Mang
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Katie Batic
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Adedeji K. Adebayo
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
- Richard L Roudebush VA Medical Center, Indianapolis, Indiana
| |
Collapse
|
24
|
Lewis MT, Caldas C. The Power and Promise of Patient-Derived Xenografts of Human Breast Cancer. Cold Spring Harb Perspect Med 2024; 14:a041329. [PMID: 38052483 PMCID: PMC10982691 DOI: 10.1101/cshperspect.a041329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
In 2016, a group of researchers engaged in the development of patient-derived xenografts (PDXs) of human breast cancer provided a comprehensive review of the state of the field. In that review, they summarized the clinical problem that PDXs might address, the technical approaches to their generation (including a discussion of host animals and transplant conditions tested), and presented transplantation success (take) rates across groups and across transplantation conditions. At the time, there were just over 500 unique PDX models created by these investigators representing all three clinically defined subtypes (ER+, HER2+, and TNBC). Today, many of these PDX resources have at least doubled in size, and several more PDX development groups now exist, such that there may be well upward of 1000 PDX models of human breast cancer in existence worldwide. They also presented a series of open questions for the field. Many of these questions have been addressed. However, several remain open, or only partially addressed. Herein, we revisit these questions, and recount the progress that has been made in a number of areas with respect to generation, characterization, and use of PDXs in translational research, and re-present questions that remain open. These open questions, and others, are now being addressed not only by individual investigators, but also large, well-funded consortia including the PDXNet program of the National Cancer Institute in the United States, and the EuroPDX Consortium, an organization of PDX developers across Europe. Finally, we discuss the new opportunities in PDX-based research.
Collapse
Affiliation(s)
- Michael T Lewis
- Baylor College of Medicine, The Lester and Sue Smith Breast Center, Departments of Molecular and Cellular Biology and Radiology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge CB2 0RE, United Kingdom
| |
Collapse
|
25
|
Lee J, Lee G, Park HS, Jeong BK, Gong G, Jeong JH, Lee HJ. Factors associated with engraftment success of patient-derived xenografts of breast cancer. Breast Cancer Res 2024; 26:49. [PMID: 38515107 PMCID: PMC10956311 DOI: 10.1186/s13058-024-01794-w] [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: 11/22/2023] [Accepted: 02/22/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Patient-derived xenograft (PDX) models serve as a valuable tool for the preclinical evaluation of novel therapies. They closely replicate the genetic, phenotypic, and histopathological characteristics of primary breast tumors. Despite their promise, the rate of successful PDX engraftment is various in the literature. This study aimed to identify the key factors associated with successful PDX engraftment of primary breast cancer. METHODS We integrated clinicopathological data with morphological attributes quantified using a trained artificial intelligence (AI) model to identify the principal factors affecting PDX engraftment. RESULTS Multivariate logistic regression analyses demonstrated that several factors, including a high Ki-67 labeling index (Ki-67LI) (p < 0.001), younger age at diagnosis (p = 0.032), post neoadjuvant chemotherapy (NAC) (p = 0.006), higher histologic grade (p = 0.039), larger tumor size (p = 0.029), and AI-assessed higher intratumoral necrosis (p = 0.027) and intratumoral invasive carcinoma (p = 0.040) proportions, were significant factors for successful PDX engraftment (area under the curve [AUC] 0.905). In the NAC group, a higher Ki-67LI (p < 0.001), lower Miller-Payne grade (p < 0.001), and reduced proportion of intratumoral normal breast glands as assessed by AI (p = 0.06) collectively provided excellent prediction accuracy for successful PDX engraftment (AUC 0.89). CONCLUSIONS We found that high Ki-67LI, younger age, post-NAC status, higher histologic grade, larger tumor size, and specific morphological attributes were significant factors for predicting successful PDX engraftment of primary breast cancer.
Collapse
Affiliation(s)
- Jongwon Lee
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - GunHee Lee
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | | | - Byung-Kwan Jeong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Gyungyub Gong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Jae Ho Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea.
| | - Hee Jin Lee
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, South Korea.
- NeogenTC Corp., Seoul, South Korea.
| |
Collapse
|
26
|
Martins-Branco D, Kassapian M, Debien V, Caparica R, Eiger D, Dafni U, Andriakopoulou C, El-Abed S, Ellard SL, Izquierdo M, Vicente M, Chumsri S, Piccart-Gebhart M, Moreno-Aspitia A, Knop AS, Lombard J, de Azambuja E. The impact of erythropoiesis-stimulating agents administration concomitantly with adjuvant anti-HER2 treatments on the outcomes of patients with early breast cancer: a sub-analysis of the ALTTO study. Breast Cancer Res Treat 2024; 203:497-509. [PMID: 37938495 PMCID: PMC11052564 DOI: 10.1007/s10549-023-07159-9] [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: 08/10/2023] [Accepted: 10/05/2023] [Indexed: 11/09/2023]
Abstract
PURPOSE To assess whether erythropoiesis-stimulating agents (ESA) administration impacts the outcomes of patients with HER2-positive early breast cancer (EBC). METHODS ALTTO (NCT00490139) patients were categorized by ESA use during adjuvant anti-HER2 treatment. Disease-free-survival (DFS), overall survival (OS), and time-to-distant recurrence (TTDR) were analyzed by ESA administration, with subgroup analyses according to prognostic factors. Log-rank tests and Cox modeling were performed. Adverse events (AEs) of ESA-interest were compared. RESULTS Among 8381 patients recruited in ALTTO, 123 (1.5%) received ESA concomitantly with study treatment. The median age of patients receiving ESA was 54 years, 39.0% premenopausal, most had tumor size > 2 cm (56.9%), node-positive (58.5%), and positive estrogen receptor expression (61.8%). Median follow-up was shorter in the ESA group [6.1 years (IQR 5.3-7.0) vs. 6.9 years (6.0-7.1); p < 0.001]. There was no DFS difference by ESA administration (log-rank p = 0.70), with 3- and 7-year DFS of 89.2% (95% CI 81.8-93.8%) and 81.6% (71.4-88.5%) in ESA group vs. 88.3% (87.6-89.0%) and 80.0% (79.1-80.9%) in No-ESA group. In subgroup analyses, the interaction of ESA administration with menopausal status was statistically significant (unadjusted p = 0.024; stratified p = 0.033), favoring premenopausal women receiving ESA. We observed no significant association of ESA administration with OS (log-rank p = 0.57; 7-year OS in ESA 88.6% vs. 90.0% in non-ESA) or TTDR. ESA-interest AEs were experienced by eight (6.5%) patients receiving ESA and 417 (5.1%) in the No-ESA group (p = 0.41). CONCLUSION ESA administration to patients receiving adjuvant anti-HER2 treatment for HER2-positive EBC was safe and not associated with a negative impact on survival outcomes.
Collapse
Affiliation(s)
- Diogo Martins-Branco
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Rue Meylemeersch, 90, Anderlecht, 1070, Brussels, Belgium
| | | | - Véronique Debien
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Rue Meylemeersch, 90, Anderlecht, 1070, Brussels, Belgium
| | - Rafael Caparica
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Rue Meylemeersch, 90, Anderlecht, 1070, Brussels, Belgium
| | - Daniel Eiger
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Rue Meylemeersch, 90, Anderlecht, 1070, Brussels, Belgium
| | - Urania Dafni
- National and Kapodistrian University of Athens & Frontier Science Foundation-Hellas, Athens, Greece
| | | | | | | | | | - Malou Vicente
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Rue Meylemeersch, 90, Anderlecht, 1070, Brussels, Belgium
| | - Saranya Chumsri
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA
| | - Martine Piccart-Gebhart
- Medical Oncology Department, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - Ann Søegaard Knop
- Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Janine Lombard
- Calvary Mater Hospital & Australia and New Zealand Breast Cancer Trials Group (BCT-ANZ), Newcastle, Australia
| | - Evandro de Azambuja
- Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), Rue Meylemeersch, 90, Anderlecht, 1070, Brussels, Belgium.
| |
Collapse
|
27
|
Upton DH, Ziegler DS, Tsoli M. Development of Orthotopic Patient-Derived Xenograft Models of Pediatric Intracranial Tumors. Methods Mol Biol 2024; 2806:75-90. [PMID: 38676797 DOI: 10.1007/978-1-0716-3858-3_7] [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: 04/29/2024]
Abstract
The development of clinically relevant and reliable models of central nervous system tumors has been instrumental in advancing the field of Neuro-Oncology. The orthotopic intracranial injection is widely used to study the growth, invasion, and spread of tumors in a controlled environment. Orthotopic models are performed to examine tumor cells isolated from a specific region in a patient in the same site or location in an animal model. Orthotopic brain tumor models are also utilized for preclinical testing of therapeutics as they closely recapitulate the behavior of such cancer and the brain environment of patients. Below, we describe our experiences in the development of murine models of pediatric brain tumors including diffuse midline glioma (DMG), glioblastoma (GBM), and medulloblastoma. The method provides an overview of intracranial stereotactic injections in mice.
Collapse
Affiliation(s)
- Dannielle H Upton
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
| | - David S Ziegler
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia.
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW, Australia.
- Kid's Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia.
| | - Maria Tsoli
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia.
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
28
|
Hanssen KM, Fletcher JI, Kamili A. Generation of Orthotopic and Subcutaneous Patient-Derived Xenograft Models from Diverse Clinical Tissue Samples of Pediatric Extracranial Solid Tumors. Methods Mol Biol 2024; 2806:55-74. [PMID: 38676796 DOI: 10.1007/978-1-0716-3858-3_6] [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: 04/29/2024]
Abstract
Realistic and renewable laboratory models that accurately reflect the distinct clinical features of childhood cancers have enormous potential to speed research progress. These models help us to understand disease biology, develop new research methods, advance new therapies to clinical trial, and implement personalized medicine. This chapter describes methods to generate patient-derived xenograft models of neuroblastoma and rhabdomyosarcoma, two tumor types for which children with high-risk disease have abysmal survival outcomes and survivors have lifelong-debilitating effects from treatment. Further, this protocol addresses model development from diverse clinical tumor tissue samples, subcutaneous and orthotopic engraftment, and approaches to avoid model loss.
Collapse
Affiliation(s)
- Kimberley M Hanssen
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
| | - Jamie I Fletcher
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
| | - Alvin Kamili
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia.
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
29
|
Feng Y, Lee J, Yang L, Hilton MB, Morris K, Seaman S, Edupuganti VVSR, Hsu KS, Dower C, Yu G, So D, Bajgain P, Zhu Z, Dimitrov DS, Patel NL, Robinson CM, Difilippantonio S, Dyba M, Corbel A, Basuli F, Swenson RE, Kalen JD, Suthe SR, Hussain M, Italia JS, Souders CA, Gao L, Schnermann MJ, St Croix B. Engineering CD276/B7-H3-targeted antibody-drug conjugates with enhanced cancer-eradicating capability. Cell Rep 2023; 42:113503. [PMID: 38019654 PMCID: PMC10872261 DOI: 10.1016/j.celrep.2023.113503] [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: 06/03/2023] [Revised: 08/18/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023] Open
Abstract
CD276/B7-H3 represents a promising target for cancer therapy based on widespread overexpression in both cancer cells and tumor-associated stroma. In previous preclinical studies, CD276 antibody-drug conjugates (ADCs) exploiting a talirine-type pyrrolobenzodiazepine (PBD) payload showed potent activity against various solid tumors but with a narrow therapeutic index and dosing regimen higher than that tolerated in clinical trials using other antibody-talirine conjugates. Here, we describe the development of a modified talirine PBD-based fully human CD276 ADC, called m276-SL-PBD, that is cross-species (human/mouse) reactive and can eradicate large 500-1,000-mm3 triple-negative breast cancer xenografts at doses 10- to 40-fold lower than the maximum tolerated dose. By combining CD276 targeting with judicious genetic and chemical ADC engineering, improved ADC purification, and payload sensitivity screening, these studies demonstrate that the therapeutic index of ADCs can be substantially increased, providing an advanced ADC development platform for potent and selective targeting of multiple solid tumor types.
Collapse
Affiliation(s)
- Yang Feng
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Jaewon Lee
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Liping Yang
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Mary Beth Hilton
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA; Basic Research Program, Frederick National Laboratory for Cancer Research (FNLCR), Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Karen Morris
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA; Basic Research Program, Frederick National Laboratory for Cancer Research (FNLCR), Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Steven Seaman
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | | | - Kuo-Sheng Hsu
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Christopher Dower
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Guojun Yu
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Daeho So
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Pradip Bajgain
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA
| | - Zhongyu Zhu
- Protein Interactions Section, Cancer and Inflammation Program, NCI, NIH, Frederick, MD 21702, USA
| | - Dimiter S Dimitrov
- Protein Interactions Section, Cancer and Inflammation Program, NCI, NIH, Frederick, MD 21702, USA
| | - Nimit L Patel
- Small Animal Imaging Program, FNLCR, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Christina M Robinson
- Animal Research Technical Support, FNLCR, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Simone Difilippantonio
- Animal Research Technical Support, FNLCR, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Marzena Dyba
- Biophysics Resource in the Center for Structural Biology, NCI, NIH, Frederick, MD, USA
| | - Amanda Corbel
- Invention Development Program, Technology Transfer Center, NCI, Frederick, MD 21701, USA
| | - Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, NIH, Rockville, MD 20850, USA
| | - Rolf E Swenson
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, NIH, Rockville, MD 20850, USA
| | - Joseph D Kalen
- Small Animal Imaging Program, FNLCR, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | | | | | | | | | - Ling Gao
- Veterans Affairs Long Beach Healthcare System, Long Beach, CA 90822, USA
| | - Martin J Schnermann
- Organic Synthesis Section, Chemical Biology Laboratory, CCR, NCI, Frederick, MD 21702, USA
| | - Brad St Croix
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Frederick, MD 21702, USA.
| |
Collapse
|
30
|
Andreani C, Bartolacci C, Persico G, Casciaro F, Amatori S, Fanelli M, Giorgio M, Galié M, Tomassoni D, Wang J, Zhang X, Bick G, Coppari R, Marchini C, Amici A. SIRT6 promotes metastasis and relapse in HER2-positive breast cancer. Sci Rep 2023; 13:22000. [PMID: 38081972 PMCID: PMC10713583 DOI: 10.1038/s41598-023-49199-7] [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: 06/09/2023] [Accepted: 12/05/2023] [Indexed: 12/18/2023] Open
Abstract
The histone deacetylase sirtuin 6 (SIRT6) has been endowed with anti-cancer capabilities in many tumor types. Here, we investigate the impact of SIRT6-overexpression (SIRT6-OE) in Delta16HER2 mice, which are a bona fide model of HER2-positive breast cancer. After an initial delay in the tumor onset, SIRT6-OE induces a more aggressive phenotype of Delta16HER2 tumors promoting the formation of higher number of tumor foci and metastases than controls. This phenotype of SIRT6-OE tumors is associated with cancer stem cell (CSC)-like features and tumor dormancy, and low senescence and oxidative DNA damage. Accordingly, a sub-set of HER2-positive breast cancer patients with concurrent SIRT6-OE has a significant poorer relapse-free survival (RFS) probability than patients with low expression of SIRT6. ChIP-seq, RNA-seq and RT-PCR experiments indicate that SIRT6-OE represses the expression of the T-box transcription factor 3 (Tbx3) by deacetylation of H3K9ac. Accordingly, loss-of-function mutations of TBX3 or low TBX3 expression levels are predictive of poor prognosis in HER2-positive breast cancer patients. Our work indicates that high levels of SIRT6 are indicative of poor prognosis and high risk of metastasis in HER2-positive breast cancer and suggests further investigation of TBX3 as a downstream target of SIRT6 and co-marker of poor-prognosis. Our results point to a breast cancer subtype-specific effect of SIRT6 and warrant future studies dissecting the mechanisms of SIRT6 regulation in different breast cancer subtypes.
Collapse
Affiliation(s)
- Cristina Andreani
- Department of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy.
- Department of Internal Medicine, University of Cincinnati, 45219, Cincinnati, OH, USA.
| | - Caterina Bartolacci
- Department of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
- Department of Internal Medicine, University of Cincinnati, 45219, Cincinnati, OH, USA
| | - Giuseppe Persico
- Department of Experimental Oncology, IRCCS-European Institute of Oncology, Via Adamello 16, 20139, Milano, Italy
| | - Francesca Casciaro
- Department of Biomedical Sciences, University of Padua, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Stefano Amatori
- Molecular Pathology Laboratory "PaoLa", Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61032, Fano, Italy
| | - Mirco Fanelli
- Molecular Pathology Laboratory "PaoLa", Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61032, Fano, Italy
| | - Marco Giorgio
- Department of Experimental Oncology, IRCCS-European Institute of Oncology, Via Adamello 16, 20139, Milano, Italy
- Department of Biomedical Sciences, University of Padua, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Mirco Galié
- Department of Neuroscience, Biomedicine and Movement, Section of Anatomy and Histology, University of Verona, 37134, Verona, Italy
| | - Daniele Tomassoni
- Department of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Junbiao Wang
- Department of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Xiaoting Zhang
- Department of Cancer Biology, University of Cincinnati, 45219, Cincinnati, OH, USA
| | - Gregory Bick
- Department of Cancer Biology, University of Cincinnati, 45219, Cincinnati, OH, USA
| | - Roberto Coppari
- Department of Cell Physiology and Metabolism, University of Geneva, 1211, Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva, 1211, Geneva, Switzerland
| | - Cristina Marchini
- Department of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy.
| | - Augusto Amici
- Department of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| |
Collapse
|
31
|
Hernández Guerrero T, Baños N, del Puerto Nevado L, Mahillo-Fernandez I, Doger De-Speville B, Calvo E, Wick M, García-Foncillas J, Moreno V. Patient Characteristics Associated with Growth of Patient-Derived Tumor Implants in Mice (Patient-Derived Xenografts). Cancers (Basel) 2023; 15:5402. [PMID: 38001663 PMCID: PMC10670531 DOI: 10.3390/cancers15225402] [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: 09/14/2023] [Revised: 10/26/2023] [Accepted: 11/04/2023] [Indexed: 11/26/2023] Open
Abstract
Background: patient-derived xenografts (PDXs) have defined the field of translational cancer research in recent years, becoming one of the most-used tools in early drug development. The process of establishing cancer models in mice has turned out to be challenging, since little research focuses on evaluating which factors impact engraftment success. We sought to determine the clinical, pathological, or molecular factors which may predict better engraftment rates in PDXs. Methods: between March 2017 and January 2021, tumor samples obtained from patients with primary or metastatic cancer were implanted into athymic nude mice. A full comprehensive evaluation of baseline factors associated with the patients and patients' tumors was performed, with the goal of potentially identifying predictive markers of engraftment. We focused on clinical (patient factors) pathological (patients' tumor samples) and molecular (patients' tumor samples) characteristics, analyzed either by immunohistochemistry (IHC) or next-generation sequencing (NGS), which were associated with the likelihood of final engraftment, as well as with tumor growth rates in xenografts. Results: a total of 585 tumor samples were collected and implanted. Twenty-one failed to engraft, due to lack of malignant cells. Of 564 tumor-positive samples, 187 (33.2%) grew at time of analysis. The study was able to find correlation and predictive value for engraftment for the following: the use of systemic antibiotics by the patient within 2 weeks of sampling (38.1% (72/189) antibiotics- group vs. 30.7% (115/375) no-antibiotics) (p = 0.048), and the administration of systemic steroids to the patients within 2 weeks of sampling (41.5% (34/48) steroids vs. 31.7% (153/329), no-steroids) (p = 0.049). Regarding patient's baseline tests, we found certain markers could help predict final engraftment success: for lactate dehydrogenase (LDH) levels, 34.1% (140/411) of tumors derived from patients with baseline blood LDH levels above the upper limit of normality (ULN) achieved growth, against 30.7% (47/153) with normal LDH (p = 0.047). Histological tumor characteristics, such as grade of differentiation, were also correlated. Grade 1: 25.4% (47/187), grade 2: 34.8% (65/187) and grade 3: 40.1% (75/187) tumors achieved successful growth (p = 0.043), suggesting the higher the grade, the higher the likelihood of success. Similarly, higher ki67 levels were also correlated with better engraftment rates: low (Ki67 < 15%): 8.9% (9/45) achieved growth vs. high (Ki67 ≥ 15%): 31% (35/113) (p: 0.002). Other markers of aggressiveness such as the presence of lymphovascular invasion in tumor sample of origin was also predictive: 42.2% (97/230) with lymphovascular vs. 26.9% (90/334) of samples with no invasion (p = 0.0001). From the molecular standpoint, mismatch-repair-deficient (MMRd) tumors showed better engraftment rates: 62.1% (18/29) achieved growth vs. 40.8% (75/184) of proficient tumors (p = 0.026). A total of 84 PDX were breast models, among which 57.9% (11/19) ER-negative models grew, vs. 15.4% (10/65) of ER-positive models (p = 0.0001), also consonant with ER-negative tumors being more aggressive. BRAFmut cancers are more likely to achieve engraftment during the development of PDX models. Lastly, tumor growth rates during first passages can help establish a cutoff point for the decision-making process during PDX development, since the higher the tumor grades, the higher the likelihood of success. Conclusions: tumors with higher grade and Ki67 protein expression, lymphovascular and/or perineural invasion, with dMMR and are negative for ER expression have a higher probability of achieving growth in the process of PDX development. The use of steroids and/or antibiotics in the patient prior to sampling can also impact the likelihood of success in PDX development. Lastly, establishing a cutoff point for tumor growth rates could guide the decision-making process during PDX development.
Collapse
Affiliation(s)
| | - Natalia Baños
- START Madrid—Fundación Jimenez Díaz University Hospital, Avenida Reyes Católicos 2, 28040 Madrid, Spain (I.M.-F.); (B.D.D.-S.); (J.G.-F.); (V.M.)
| | | | - Ignacio Mahillo-Fernandez
- START Madrid—Fundación Jimenez Díaz University Hospital, Avenida Reyes Católicos 2, 28040 Madrid, Spain (I.M.-F.); (B.D.D.-S.); (J.G.-F.); (V.M.)
- Translational Oncology Division, IIS-Fundación Jiménez Díaz-UAM, 28040 Madrid, Spain;
| | - Bernard Doger De-Speville
- START Madrid—Fundación Jimenez Díaz University Hospital, Avenida Reyes Católicos 2, 28040 Madrid, Spain (I.M.-F.); (B.D.D.-S.); (J.G.-F.); (V.M.)
| | - Emiliano Calvo
- START Madrid—CIOCC HM Sanchinarro, C. de Oña, 10, 28050 Madrid, Spain;
| | - Michael Wick
- XENOStart START San Antonio, 4383 Medical Dr, San Antonio, TX 78229, USA;
| | - Jesús García-Foncillas
- START Madrid—Fundación Jimenez Díaz University Hospital, Avenida Reyes Católicos 2, 28040 Madrid, Spain (I.M.-F.); (B.D.D.-S.); (J.G.-F.); (V.M.)
- Translational Oncology Division, IIS-Fundación Jiménez Díaz-UAM, 28040 Madrid, Spain;
| | - Victor Moreno
- START Madrid—Fundación Jimenez Díaz University Hospital, Avenida Reyes Católicos 2, 28040 Madrid, Spain (I.M.-F.); (B.D.D.-S.); (J.G.-F.); (V.M.)
| |
Collapse
|
32
|
Chen HK, Chen YL, Wang CY, Chung WP, Fang JH, Lai MD, Hsu HP. ABCB1 Regulates Immune Genes in Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:801-811. [PMID: 38020048 PMCID: PMC10655737 DOI: 10.2147/bctt.s421213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023]
Abstract
Background Resistance to standard chemotherapy is a critical problem for breast cancer patients. The ATP-binding cassette (ABC) superfamily transporters actively pump out drugs and play an important role in chemoresistance. ABCB1 (ABC subfamily B, member 1, also named as multidrug resistance protein 1, MDR1) and suppressive myeloid-derived suppressor cells (MDSCs) potentially involve in chemoresistance of breast cancer. The relationship between ABCB1 and immune genes in breast cancer has not been widely studied. Methods Microarray and RNA sequencing data were obtained from The Cancer Genome Atlas Breast Invasive Carcinoma in Genomic Data Commons Data Portal and Gene Expression Omnibus database. A patient-derived xenograft (PDX) model of HER2+ breast cancer was established to investigate the association between ABCB1 and immune genes in breast cancer. Results Expression of ABCB1 increased in doxorubicin-selected MCF-7/ADR cells. High expression of ABCB1 mRNA is correlated with lymph-node metastasis and worse overall survival in patients with breast cancer. ABCB1 is positively correlated with IL6, CSF1, CSF3, and PTGS2. In the HER2+ stage IIA breast cancer PDX model, both doxorubicin and paclitaxel suppressed growth of P2 tumors. IL6, CSF1, CSF3, and PTGS2 expression were suppressed by paclitaxel but not doxorubicin. Intrasplenic MDSCs, including CD11b+Ly6G+ and CD11b+Ly6C+ cells, were more abundant than intratumor MDSCs in PDX-carrying nude mice. Clinically, the patient developed cancer recurrence after adjuvant chemotherapy with doxorubicin-based regimen and was well controlled after paclitaxel-trastuzumab combined therapy. Conclusion ABCB1 was a poor predictor of HER2+ LN- breast cancer. Regulation of immune genes by ABCB1 contributed to cancer recurrence and treatment effect. The PDX model was suitable for investigation the expression of target genes and expansion of immune cells.
Collapse
Affiliation(s)
- Han-Kun Chen
- Department of Surgery, Chi Mei Medical Center, Tainan, Taiwan
| | - Yi-Ling Chen
- Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Chih-Yang Wang
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Wei-Pang Chung
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70403, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, Taiwan
| | - Jung-Hua Fang
- Laboratory Animal Center, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ming-Derg Lai
- Department of Biochemistry and Molecular Biology, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hui-Ping Hsu
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
33
|
Lin FT, Liu K, Garan LAW, Folly-Kossi H, Song Y, Lin SJ, Lin WC. A small-molecule inhibitor of TopBP1 exerts anti-MYC activity and synergy with PARP inhibitors. Proc Natl Acad Sci U S A 2023; 120:e2307793120. [PMID: 37878724 PMCID: PMC10622895 DOI: 10.1073/pnas.2307793120] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/19/2023] [Indexed: 10/27/2023] Open
Abstract
We have previously identified TopBP1 (topoisomerase IIβ-binding protein 1) as a promising target for cancer therapy, given its role in the convergence of Rb, PI(3)K/Akt, and p53 pathways. Based on this, we conducted a large-scale molecular docking screening to identify a small-molecule inhibitor that specifically targets the BRCT7/8 domains of TopBP1, which we have named 5D4. Our studies show that 5D4 inhibits TopBP1 interactions with E2F1, mutant p53, and Cancerous Inhibitor of Protein Phosphatase 2A. This leads to the activation of E2F1-mediated apoptosis and the inhibition of mutant p53 gain of function. In addition, 5D4 disrupts the interaction of TopBP1 with MIZ1, which in turn allows MIZ1 to bind to its target gene promoters and repress MYC activity. Moreover, 5D4 inhibits the association of the TopBP1-PLK1 complex and prevents the formation of Rad51 foci. When combined with inhibitors of PARP1/2 or PARP14, 5D4 synergizes to effectively block cancer cell proliferation. Our animal studies have demonstrated the antitumor activity of 5D4 in breast and ovarian cancer xenograft models. Moreover, the effectiveness of 5D4 is further enhanced when combined with a PARP1/2 inhibitor talazoparib. Taken together, our findings strongly support the potential use of TopBP1-BRCT7/8 inhibitors as a targeted cancer therapy.
Collapse
Affiliation(s)
- Fang-Tsyr Lin
- Section of Hematology/Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX77030
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX77030
| | - Kang Liu
- Section of Hematology/Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX77030
| | - Lidija A. Wilhelms Garan
- Section of Hematology/Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX77030
- Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, TX77030
| | - Helena Folly-Kossi
- Section of Hematology/Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX77030
| | - Yongcheng Song
- Department of Pharmacology, Baylor College of Medicine, Houston, TX77030
| | - Shwu-Jiuan Lin
- Department of Pharmaceutical Sciences, School of Pharmacy, Taipei Medical University, Taipei11031, Taiwan
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| | - Weei-Chin Lin
- Section of Hematology/Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX77030
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX77030
- Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, TX77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX77030
| |
Collapse
|
34
|
Gou X, Kim BJ, Anurag M, Lei JT, Young MN, Holt MV, Fandino D, Vollert CT, Singh P, Alzubi MA, Malovannaya A, Dobrolecki LE, Lewis MT, Li S, Foulds CE, Ellis MJ. Kinome Reprogramming Is a Targetable Vulnerability in ESR1 Fusion-Driven Breast Cancer. Cancer Res 2023; 83:3237-3251. [PMID: 37071495 PMCID: PMC10543968 DOI: 10.1158/0008-5472.can-22-3484] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/20/2023] [Accepted: 04/12/2023] [Indexed: 04/19/2023]
Abstract
Transcriptionally active ESR1 fusions (ESR1-TAF) are a potent cause of breast cancer endocrine therapy (ET) resistance. ESR1-TAFs are not directly druggable because the C-terminal estrogen/anti-estrogen-binding domain is replaced with translocated in-frame partner gene sequences that confer constitutive transactivation. To discover alternative treatments, a mass spectrometry (MS)-based kinase inhibitor pulldown assay (KIPA) was deployed to identify druggable kinases that are upregulated by diverse ESR1-TAFs. Subsequent explorations of drug sensitivity validated RET kinase as a common therapeutic vulnerability despite remarkable ESR1-TAF C-terminal sequence and structural diversity. Organoids and xenografts from a pan-ET-resistant patient-derived xenograft model that harbors the ESR1-e6>YAP1 TAF were concordantly inhibited by the selective RET inhibitor pralsetinib to a similar extent as the CDK4/6 inhibitor palbociclib. Together, these findings provide preclinical rationale for clinical evaluation of RET inhibition for the treatment of ESR1-TAF-driven ET-resistant breast cancer. SIGNIFICANCE Kinome analysis of ESR1 translocated and mutated breast tumors using drug bead-based mass spectrometry followed by drug-sensitivity studies nominates RET as a therapeutic target. See related commentary by Wu and Subbiah, p. 3159.
Collapse
Affiliation(s)
- Xuxu Gou
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston Texas
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Beom-Jun Kim
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Meenakshi Anurag
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Jonathan T. Lei
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, Texas
| | - Meggie N. Young
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas
| | - Matthew V. Holt
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Diana Fandino
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Craig T. Vollert
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Employee of Adrienne Helis Malvin Medical Research Foundation, New Orleans, Los Angeles
| | - Purba Singh
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Mohammad A. Alzubi
- Employee of Adrienne Helis Malvin Medical Research Foundation, New Orleans, Los Angeles
| | - Anna Malovannaya
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas
| | - Lacey E. Dobrolecki
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Michael T. Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Radiology, Baylor College of Medicine, Houston, Texas
| | - Shunqiang Li
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Charles E. Foulds
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Matthew J. Ellis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston Texas
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
35
|
Daneshdoust D, Luo M, Li Z, Mo X, Alothman S, Kallakury B, Schlegel R, Zhang J, Guo D, Furth PA, Liu X, Li J. Unlocking Translational Potential: Conditionally Reprogrammed Cells in Advancing Breast Cancer Research. Cells 2023; 12:2388. [PMID: 37830602 PMCID: PMC10572051 DOI: 10.3390/cells12192388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/07/2023] [Accepted: 09/19/2023] [Indexed: 10/14/2023] Open
Abstract
Preclinical in vitro models play an important role in studying cancer cell biology and facilitating translational research, especially in the identification of drug targets and drug discovery studies. This is particularly relevant in breast cancer, where the global burden of disease is quite high based on prevalence and a relatively high rate of lethality. Predictive tools to select patients who will be responsive to invasive or morbid therapies (radiotherapy, chemotherapy, immunotherapy, and/or surgery) are relatively lacking. To be clinically relevant, a model must accurately replicate the biology and cellular heterogeneity of the primary tumor. Addressing these requirements and overcoming the limitations of most existing cancer cell lines, which are typically derived from a single clone, we have recently developed conditional reprogramming (CR) technology. The CR technology refers to a co-culture system of primary human normal or tumor cells with irradiated murine fibroblasts in the presence of a Rho-associated kinase inhibitor to allow the primary cells to acquire stem cell properties and the ability to proliferate indefinitely in vitro without any exogenous gene or viral transfection. This innovative approach fulfills many of these needs and offers an alternative that surpasses the deficiencies associated with traditional cancer cell lines. These CR cells (CRCs) can be reprogrammed to maintain a highly proliferative state and reproduce the genomic and histological characteristics of the parental tissue. Therefore, CR technology may be a clinically relevant model to test and predict drug sensitivity, conduct gene profile analysis and xenograft research, and undertake personalized medicine. This review discusses studies that have applied CR technology to conduct breast cancer research.
Collapse
Affiliation(s)
- Danyal Daneshdoust
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Mingjue Luo
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| | - Zaibo Li
- Departments of Pathology, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA
| | - Xiaokui Mo
- Department of Biostatics and Bioinformatics, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA
| | - Sahar Alothman
- Departments of Oncology and Medicine, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Bhaskar Kallakury
- Departments of Pathology, Lombardi Comprehensive Cancer Center, Center for Cell Reprogramming, Georgetown University, Washington, DC 20057, USA
| | - Richard Schlegel
- Departments of Pathology, Lombardi Comprehensive Cancer Center, Center for Cell Reprogramming, Georgetown University, Washington, DC 20057, USA
| | - Junran Zhang
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
- Department of Radiation Oncology, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA
| | - Deliang Guo
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
- Department of Radiation Oncology, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA
| | - Priscilla A. Furth
- Departments of Oncology and Medicine, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Xuefeng Liu
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
- Departments of Pathology, Urology, and Radiation Oncology, Wexner Medical Center, Ohio State University, Columbus, OH 43210, USA
| | - Jenny Li
- Comprehensive Cancer Center, Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
36
|
Liu L, Wu M, Huang A, Gao C, Yang Y, Liu H, Jiang H, Yu L, Huang Y, Wang H. Establishment of a high-fidelity patient-derived xenograft model for cervical cancer enables the evaluation of patient's response to conventional and novel therapies. J Transl Med 2023; 21:611. [PMID: 37689699 PMCID: PMC10492358 DOI: 10.1186/s12967-023-04444-5] [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: 06/08/2023] [Accepted: 08/16/2023] [Indexed: 09/11/2023] Open
Abstract
BACKGROUND Recurrent or metastatic cervical cancer (r/m CC) often has poor prognosis owing to its limited treatment options. The development of novel therapeutic strategies has been hindered by the lack of preclinical models that accurately reflect the biological and genomic heterogeneity of cervical cancer (CC). Herein, we aimed to establish a large patient-derived xenograft (PDX) biobank for CC, evaluate the consistency of the biologic indicators between PDX and primary tumor tissues of patients, and explore its utility for assessing patient's response to conventional and novel therapies. METHODS Sixty-nine fresh CC tumor tissues were implanted directly into immunodeficient mice to establish PDX models. The concordance of the PDX models with their corresponding primary tumors (PTs) was compared based on the clinical pathological features, protein biomarker levels, and genomic features through hematoxylin & eosin staining, immunohistochemistry, and whole exome sequencing, respectively. Moreover, the clinical information of CC patients, RNA transcriptome and immune phenotyping of primary tumors were integrated to identify the potential parameters that could affect the success of xenograft engraftment. Subsequently, PDX model was evaluated for its capacity to mirror patient's response to chemotherapy. Finally, PDX model and PDX-derived organoid (PDXO) were utilized to evaluate the therapeutic efficacy of neratinib and adoptive cell therapy (ACT) combination strategy for CC patients with human epidermal growth factor receptor 2 (HER2) mutation. RESULTS We established a PDX biobank for CC with a success rate of 63.8% (44/69). The primary features of established PDX tumors, including clinicopathological features, the expression levels of protein biomarkers including Ki67, α-smooth muscle actin, and p16, and genomics, were highly consistent with their PTs. Furthermore, xenograft engraftment was likely influenced by the primary tumor size, the presence of follicular helper T cells and the expression of cell adhesion-related genes in primary tumor tissue. The CC derived PDX models were capable of recapitulating the patient's response to chemotherapy. In a PDX model, a novel therapeutic strategy, the combination of ACT and neratinib, was shown to effectively inhibit the growth of PDX tumors derived from CC patients with HER2-mutation. CONCLUSIONS We established by far the largest PDX biobank with a high engraftment rate for CC that preserves the histopathological and genetic characteristics of patient's biopsy samples, recapitulates patient's response to conventional therapy, and is capable of evaluating the efficacy of novel therapeutic modalities for CC.
Collapse
Affiliation(s)
- Liting Liu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anni Huang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chun Gao
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yifan Yang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Liu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Jiang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Long Yu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yafei Huang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Hui Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
37
|
Yao TH, Wu Z, Bharath K, Li J, Baladandayuthapani V. PROBABILISTIC LEARNING OF TREATMENT TREES IN CANCER. Ann Appl Stat 2023; 17:1884-1908. [PMID: 37711665 PMCID: PMC10501503 DOI: 10.1214/22-aoas1696] [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] [Indexed: 09/16/2023]
Abstract
Accurate identification of synergistic treatment combinations and their underlying biological mechanisms is critical across many disease domains, especially cancer. In translational oncology research, preclinical systems such as patient-derived xenografts (PDX) have emerged as a unique study design evaluating multiple treatments administered to samples from the same human tumor implanted into genetically identical mice. In this paper, we propose a novel Bayesian probabilistic tree-based framework for PDX data to investigate the hierarchical relationships between treatments by inferring treatment cluster trees, referred to as treatment trees (Rx-tree). The framework motivates a new metric of mechanistic similarity between two or more treatments accounting for inherent uncertainty in tree estimation; treatments with a high estimated similarity have potentially high mechanistic synergy. Building upon Dirichlet Diffusion Trees, we derive a closed-form marginal likelihood encoding the tree structure, which facilitates computationally efficient posterior inference via a new two-stage algorithm. Simulation studies demonstrate superior performance of the proposed method in recovering the tree structure and treatment similarities. Our analyses of a recently collated PDX dataset produce treatment similarity estimates that show a high degree of concordance with known biological mechanisms across treatments in five different cancers. More importantly, we uncover new and potentially effective combination therapies that confer synergistic regulation of specific downstream biological pathways for future clinical investigations. Our accompanying code, data, and shiny application for visualization of results are available at: https://github.com/bayesrx/RxTree.
Collapse
Affiliation(s)
- Tsung-Hung Yao
- Department of Biostatistics, University of Michigan at Ann Arbor
| | - Zhenke Wu
- Department of Biostatistics, University of Michigan at Ann Arbor
| | | | - Jinju Li
- Department of Biostatistics, University of Michigan at Ann Arbor
| | | |
Collapse
|
38
|
Zeng M, Ruan Z, Tang J, Liu M, Hu C, Fan P, Dai X. Generation, evolution, interfering factors, applications, and challenges of patient-derived xenograft models in immunodeficient mice. Cancer Cell Int 2023; 23:120. [PMID: 37344821 DOI: 10.1186/s12935-023-02953-3] [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: 03/09/2023] [Accepted: 05/24/2023] [Indexed: 06/23/2023] Open
Abstract
Establishing appropriate preclinical models is essential for cancer research. Evidence suggests that cancer is a highly heterogeneous disease. This follows the growing use of cancer models in cancer research to avoid these differences between xenograft tumor models and patient tumors. In recent years, a patient-derived xenograft (PDX) tumor model has been actively generated and applied, which preserves both cell-cell interactions and the microenvironment of tumors by directly transplanting cancer tissue from tumors into immunodeficient mice. In addition to this, the advent of alternative hosts, such as zebrafish hosts, or in vitro models (organoids and microfluidics), has also facilitated the advancement of cancer research. However, they still have a long way to go before they become reliable models. The development of immunodeficient mice has enabled PDX to become more mature and radiate new vitality. As one of the most reliable and standard preclinical models, the PDX model in immunodeficient mice (PDX-IM) exerts important effects in drug screening, biomarker development, personalized medicine, co-clinical trials, and immunotherapy. Here, we focus on the development procedures and application of PDX-IM in detail, summarize the implications that the evolution of immunodeficient mice has brought to PDX-IM, and cover the key issues in developing PDX-IM in preclinical studies.
Collapse
Affiliation(s)
- Mingtang Zeng
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zijing Ruan
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiaxi Tang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Maozhu Liu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chengji Hu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ping Fan
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Xinhua Dai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
39
|
Stashko C, Hayward MK, Northey JJ, Pearson N, Ironside AJ, Lakins JN, Oria R, Goyette MA, Mayo L, Russnes HG, Hwang ES, Kutys ML, Polyak K, Weaver VM. A convolutional neural network STIFMap reveals associations between stromal stiffness and EMT in breast cancer. Nat Commun 2023; 14:3561. [PMID: 37322009 PMCID: PMC10272194 DOI: 10.1038/s41467-023-39085-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 05/26/2023] [Indexed: 06/17/2023] Open
Abstract
Intratumor heterogeneity associates with poor patient outcome. Stromal stiffening also accompanies cancer. Whether cancers demonstrate stiffness heterogeneity, and if this is linked to tumor cell heterogeneity remains unclear. We developed a method to measure the stiffness heterogeneity in human breast tumors that quantifies the stromal stiffness each cell experiences and permits visual registration with biomarkers of tumor progression. We present Spatially Transformed Inferential Force Map (STIFMap) which exploits computer vision to precisely automate atomic force microscopy (AFM) indentation combined with a trained convolutional neural network to predict stromal elasticity with micron-resolution using collagen morphological features and ground truth AFM data. We registered high-elasticity regions within human breast tumors colocalizing with markers of mechanical activation and an epithelial-to-mesenchymal transition (EMT). The findings highlight the utility of STIFMap to assess mechanical heterogeneity of human tumors across length scales from single cells to whole tissues and implicates stromal stiffness in tumor cell heterogeneity.
Collapse
Affiliation(s)
- Connor Stashko
- Department of Surgery, University of California, San Francisco, CA, USA
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA
| | - Mary-Kate Hayward
- Department of Surgery, University of California, San Francisco, CA, USA
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA
| | - Jason J Northey
- Department of Surgery, University of California, San Francisco, CA, USA
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA
| | | | - Alastair J Ironside
- Department of Pathology, Western General Hospital, NHS Lothian, Edinburgh, UK
| | - Johnathon N Lakins
- Department of Surgery, University of California, San Francisco, CA, USA
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA
| | - Roger Oria
- Department of Surgery, University of California, San Francisco, CA, USA
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA
| | - Marie-Anne Goyette
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Lakyn Mayo
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
| | - Hege G Russnes
- Department of Pathology and Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - E Shelley Hwang
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Matthew L Kutys
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Valerie M Weaver
- Department of Surgery, University of California, San Francisco, CA, USA.
- Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, USA.
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
- Department of Radiation Oncology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
40
|
Mundi PS, Dela Cruz FS, Grunn A, Diolaiti D, Mauguen A, Rainey AR, Guillan K, Siddiquee A, You D, Realubit R, Karan C, Ortiz MV, Douglass EF, Accordino M, Mistretta S, Brogan F, Bruce JN, Caescu CI, Carvajal RD, Crew KD, Decastro G, Heaney M, Henick BS, Hershman DL, Hou JY, Iwamoto FM, Jurcic JG, Kiran RP, Kluger MD, Kreisl T, Lamanna N, Lassman AB, Lim EA, Manji GA, McKhann GM, McKiernan JM, Neugut AI, Olive KP, Rosenblat T, Schwartz GK, Shu CA, Sisti MB, Tergas A, Vattakalam RM, Welch M, Wenske S, Wright JD, Hibshoosh H, Kalinsky K, Aburi M, Sims PA, Alvarez MJ, Kung AL, Califano A. A Transcriptome-Based Precision Oncology Platform for Patient-Therapy Alignment in a Diverse Set of Treatment-Resistant Malignancies. Cancer Discov 2023; 13:1386-1407. [PMID: 37061969 PMCID: PMC10239356 DOI: 10.1158/2159-8290.cd-22-1020] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 01/14/2023] [Accepted: 03/14/2023] [Indexed: 04/17/2023]
Abstract
Predicting in vivo response to antineoplastics remains an elusive challenge. We performed a first-of-kind evaluation of two transcriptome-based precision cancer medicine methodologies to predict tumor sensitivity to a comprehensive repertoire of clinically relevant oncology drugs, whose mechanism of action we experimentally assessed in cognate cell lines. We enrolled patients with histologically distinct, poor-prognosis malignancies who had progressed on multiple therapies, and developed low-passage, patient-derived xenograft models that were used to validate 35 patient-specific drug predictions. Both OncoTarget, which identifies high-affinity inhibitors of individual master regulator (MR) proteins, and OncoTreat, which identifies drugs that invert the transcriptional activity of hyperconnected MR modules, produced highly significant 30-day disease control rates (68% and 91%, respectively). Moreover, of 18 OncoTreat-predicted drugs, 15 induced the predicted MR-module activity inversion in vivo. Predicted drugs significantly outperformed antineoplastic drugs selected as unpredicted controls, suggesting these methods may substantively complement existing precision cancer medicine approaches, as also illustrated by a case study. SIGNIFICANCE Complementary precision cancer medicine paradigms are needed to broaden the clinical benefit realized through genetic profiling and immunotherapy. In this first-in-class application, we introduce two transcriptome-based tumor-agnostic systems biology tools to predict drug response in vivo. OncoTarget and OncoTreat are scalable for the design of basket and umbrella clinical trials. This article is highlighted in the In This Issue feature, p. 1275.
Collapse
Affiliation(s)
- Prabhjot S. Mundi
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
| | - Filemon S. Dela Cruz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY USA 10065
| | - Adina Grunn
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
| | - Daniel Diolaiti
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY USA 10065
| | - Audrey Mauguen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY USA 10065
| | - Allison R. Rainey
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY USA 10065
| | - Kristina Guillan
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY USA 10065
| | - Armaan Siddiquee
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY USA 10065
| | - Daoqi You
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY USA 10065
| | - Ronald Realubit
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
| | - Charles Karan
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
| | - Michael V. Ortiz
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY USA 10065
| | - Eugene F. Douglass
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
| | - Melissa Accordino
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Suzanne Mistretta
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
| | - Frances Brogan
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
| | - Jeffrey N. Bruce
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Neurological Surgery, Columbia University Irving Medical Center, 710 W 168th Street, New York, NY USA 10032
| | - Cristina I. Caescu
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
| | - Richard D. Carvajal
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Katherine D Crew
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Guarionex Decastro
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Urology, Columbia University Irving Medical Center, 160 Fort Washington Ave, New York, NY USA 10032
| | - Mark Heaney
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Brian S Henick
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Dawn L Hershman
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
- Department of Epidemiology, Columbia University Mailman School of Public Health, 722 West 168th St. NY, NY 10032
| | - June Y. Hou
- Department of Obstetrics & Gynecology, Columbia University Irving Medical Center, 622 W 168th Street, New York, NY USA 10032
| | - Fabio M. Iwamoto
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Neurology, Columbia University Irving Medical Center, 710 W 168th Street, New York, NY USA 10032
| | - Joseph G. Jurcic
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Ravi P. Kiran
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Surgery, Columbia University Irving Medical Center, 622 W 168th Street, New York, NY USA 10032
| | - Michael D Kluger
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Surgery, Columbia University Irving Medical Center, 622 W 168th Street, New York, NY USA 10032
| | - Teri Kreisl
- Novartis Five Cambridge, MA 02142, United States
| | - Nicole Lamanna
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Andrew B. Lassman
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Neurology, Columbia University Irving Medical Center, 710 W 168th Street, New York, NY USA 10032
| | - Emerson A. Lim
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Gulam A. Manji
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Guy M McKhann
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Neurological Surgery, Columbia University Irving Medical Center, 710 W 168th Street, New York, NY USA 10032
| | - James M. McKiernan
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Urology, Columbia University Irving Medical Center, 160 Fort Washington Ave, New York, NY USA 10032
| | - Alfred I Neugut
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
- Department of Epidemiology, Columbia University Mailman School of Public Health, 722 West 168th St. NY, NY 10032
| | - Kenneth P. Olive
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Todd Rosenblat
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Gary K. Schwartz
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Catherine A Shu
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Michael B. Sisti
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Neurological Surgery, Columbia University Irving Medical Center, 710 W 168th Street, New York, NY USA 10032
- Department of Otolaryngology Head and Neck Surgery, Columbia University Irving Medical Center, 710 W 168th Street, New York, NY USA 10032
- Department of Radiation Oncology, Columbia University Irving Medical Center, 161 Fort Washington Avenue, New York, NY 10032, United States
| | - Ana Tergas
- Department of Obstetrics & Gynecology, Columbia University Irving Medical Center, 622 W 168th Street, New York, NY USA 10032
| | - Reena M Vattakalam
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Obstetrics & Gynecology, Columbia University Irving Medical Center, 622 W 168th Street, New York, NY USA 10032
| | - Mary Welch
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Neurology, Columbia University Irving Medical Center, 710 W 168th Street, New York, NY USA 10032
| | - Sven Wenske
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Urology, Columbia University Irving Medical Center, 160 Fort Washington Ave, New York, NY USA 10032
| | - Jason D. Wright
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Obstetrics & Gynecology, Columbia University Irving Medical Center, 622 W 168th Street, New York, NY USA 10032
| | - Hanina Hibshoosh
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
| | - Kevin Kalinsky
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Winship Cancer Institute of Emory University and Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365-C Clifton Road NE, Atlanta, GA 30322, United States
| | - Mahalaxmi Aburi
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
| | - Peter A. Sims
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Biochemistry & Molecular Biophysics, Columbia University Irving Medical Center, 701 W 168th Street, New York, NY USA 10032
| | - Mariano J. Alvarez
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- DarwinHealth Inc. New York
| | - Andrew L. Kung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY USA 10065
| | - Andrea Califano
- Department of Systems Biology, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, 1130 Saint Nicholas Ave, New York, NY USA 10032
- Department of Medicine, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY USA 10032
- Department of Biochemistry & Molecular Biophysics, Columbia University Irving Medical Center, 701 W 168th Street, New York, NY USA 10032
- Department of Biomedical Informatics, Columbia University Irving Medical Center, 622 W 168th Street, New York, NY USA 10032
- J.P. Sulzberger Columbia Genome Center, Columbia University Irving Medical Center, 622 W 168th Street, New York, NY USA 10032
| |
Collapse
|
41
|
Chen A, Neuwirth I, Herndler-Brandstetter D. Modeling the Tumor Microenvironment and Cancer Immunotherapy in Next-Generation Humanized Mice. Cancers (Basel) 2023; 15:2989. [PMID: 37296949 PMCID: PMC10251926 DOI: 10.3390/cancers15112989] [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: 03/13/2023] [Revised: 05/10/2023] [Accepted: 05/28/2023] [Indexed: 06/12/2023] Open
Abstract
Cancer immunotherapy has brought significant clinical benefits to numerous patients with malignant disease. However, only a fraction of patients experiences complete and durable responses to currently available immunotherapies. This highlights the need for more effective immunotherapies, combination treatments and predictive biomarkers. The molecular properties of a tumor, intratumor heterogeneity and the tumor immune microenvironment decisively shape tumor evolution, metastasis and therapy resistance and are therefore key targets for precision cancer medicine. Humanized mice that support the engraftment of patient-derived tumors and recapitulate the human tumor immune microenvironment of patients represent a promising preclinical model to address fundamental questions in precision immuno-oncology and cancer immunotherapy. In this review, we provide an overview of next-generation humanized mouse models suitable for the establishment and study of patient-derived tumors. Furthermore, we discuss the opportunities and challenges of modeling the tumor immune microenvironment and testing a variety of immunotherapeutic approaches using human immune system mouse models.
Collapse
Affiliation(s)
| | | | - Dietmar Herndler-Brandstetter
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, 1090 Vienna, Austria; (A.C.); (I.N.)
| |
Collapse
|
42
|
Salemme V, Centonze G, Avalle L, Natalini D, Piccolantonio A, Arina P, Morellato A, Ala U, Taverna D, Turco E, Defilippi P. The role of tumor microenvironment in drug resistance: emerging technologies to unravel breast cancer heterogeneity. Front Oncol 2023; 13:1170264. [PMID: 37265795 PMCID: PMC10229846 DOI: 10.3389/fonc.2023.1170264] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/28/2023] [Indexed: 06/03/2023] Open
Abstract
Breast cancer is a highly heterogeneous disease, at both inter- and intra-tumor levels, and this heterogeneity is a crucial determinant of malignant progression and response to treatments. In addition to genetic diversity and plasticity of cancer cells, the tumor microenvironment contributes to tumor heterogeneity shaping the physical and biological surroundings of the tumor. The activity of certain types of immune, endothelial or mesenchymal cells in the microenvironment can change the effectiveness of cancer therapies via a plethora of different mechanisms. Therefore, deciphering the interactions between the distinct cell types, their spatial organization and their specific contribution to tumor growth and drug sensitivity is still a major challenge. Dissecting intra-tumor heterogeneity is currently an urgent need to better define breast cancer biology and to develop therapeutic strategies targeting the microenvironment as helpful tools for combined and personalized treatment. In this review, we analyze the mechanisms by which the tumor microenvironment affects the characteristics of tumor heterogeneity that ultimately result in drug resistance, and we outline state of the art preclinical models and emerging technologies that will be instrumental in unraveling the impact of the tumor microenvironment on resistance to therapies.
Collapse
Affiliation(s)
- Vincenzo Salemme
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Giorgia Centonze
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Lidia Avalle
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Dora Natalini
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Alessio Piccolantonio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Pietro Arina
- UCL, Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, United Kingdom
| | - Alessandro Morellato
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Ugo Ala
- Department of Veterinary Sciences, University of Turin, Grugliasco, TO, Italy
| | - Daniela Taverna
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Paola Defilippi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
- Molecular Biotechnology Center (MBC) “Guido Tarone”, Turin, Italy
| |
Collapse
|
43
|
Li R, Mukherjee MB, Jin Z, Liu H, Lin K, Liu Q, Dilger JP, Lin J. The Potential Effect of General Anesthetics in Cancer Surgery: Meta-Analysis of Postoperative Metastasis and Inflammatory Cytokines. Cancers (Basel) 2023; 15:2759. [PMID: 37345096 PMCID: PMC10216624 DOI: 10.3390/cancers15102759] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 05/01/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023] Open
Abstract
Metastasis or recurrence following curative surgery is the main indicator of tumor progress and is the main cause of patient death. For more than three decades, the potential for general anesthesia to affect cancer outcomes has been a subject of concern with considerable research interest. Here, we conducted this systematic review and meta-analysis to summarize the effect of inhalational anesthesia (IHNA) vs. propofol-based total intravenous anesthesia (TIVA) on metastasis and recurrence after cancer surgery from clinical and pre-clinical studies. The relative risk for metastasis/recurrence in TIVA is 0.61 (95% confidence interval (95% CI) 0.46 to 0.82, p = 0.0009) compared to IHNA. Inflammatory cytokines have been implicated in cancer metastasis following cancer surgery, thus we analyzed inflammatory cytokines levels after surgery under IHNA or TIVA. Based on pooled analysis, a lower IL-6 level was noticed in TIVA in comparison to IHNA (standardized mean difference (SMD) = 0.77, 95% CI = 0.097 to 1.44, I2 = 92%, p = 0.02) but not TNF-α or IL-10. Preclinical animal model studies show that inhalational anesthetics increase the risk of breast cancer metastasis compared to propofol. In conclusion, the current evidence suggests intravenous anesthetic propofol is associated with less metastasis/recurrence and lower postoperative IL-6 level over inhaled anesthetics in the oncological surgery. We urge more well-designed clinical and preclinical studies in this field.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jun Lin
- Department of Anesthesiology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY 11794-8480, USA
| |
Collapse
|
44
|
Massicano AVF, Song PN, Mansur A, White SL, Sorace AG, Lapi SE. [ 89Zr]-Atezolizumab-PET Imaging Reveals Longitudinal Alterations in PDL1 during Therapy in TNBC Preclinical Models. Cancers (Basel) 2023; 15:2708. [PMID: 37345044 PMCID: PMC10216761 DOI: 10.3390/cancers15102708] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 06/23/2023] Open
Abstract
Triple-negative breast cancers (TNBCs) currently have limited treatment options; however, PD-L1 is an indicator of susceptibility to immunotherapy. Currently, assessment of PD-L1 is limited to biopsy samples. These limitations may be overcome with molecular imaging. In this work, we describe chemistry development and optimization, in vitro, in vivo, and dosimetry of [89Zr]-Atezolizumab for PD-L1 imaging. Atezolizumab was conjugated to DFO and radiolabeled with 89Zr. Tumor uptake and heterogeneity in TNBC xenograft and patient-derived xenograft (PDX) mouse models were quantified following [89Zr]-Atezolizumab-PET imaging. PD-L1 expression in TNBC PDX models undergoing therapy and immunohistochemistry (IHC) was used to validate imaging. SUV from PET imaging was quantified and used to identify heterogeneity. PET/CT imaging using [89Zr]-Atezolizumab identified a significant increase in tumor:muscle SUVmean 1 and 4 days after niraparib therapy and revealed an increased trend in PD-L1 expression following other cytotoxic therapies. A preliminary dosimetry study indicated the organs that will receive a higher dose are the spleen, adrenals, kidneys, and liver. [89Zr]-Atezolizumab PET/CT imaging reveals potential for the noninvasive detection of PD-L1-positive TNBC tumors and allows for quantitative and longitudinal assessment. This has potential significance for understanding tumor heterogeneity and monitoring early expression changes in PD-L1 induced by therapy.
Collapse
Affiliation(s)
| | - Patrick N. Song
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Department of Graduate Biomedical Sciences, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Ameer Mansur
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Sharon L. White
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Anna G. Sorace
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Suzanne E. Lapi
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| |
Collapse
|
45
|
Manduca N, Maccafeo E, De Maria R, Sistigu A, Musella M. 3D cancer models: One step closer to in vitro human studies. Front Immunol 2023; 14:1175503. [PMID: 37114038 PMCID: PMC10126361 DOI: 10.3389/fimmu.2023.1175503] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/23/2023] [Indexed: 04/29/2023] Open
Abstract
Cancer immunotherapy is the great breakthrough in cancer treatment as it displayed prolonged progression-free survival over conventional therapies, yet, to date, in only a minority of patients. In order to broad cancer immunotherapy clinical applicability some roadblocks need to be overcome, first among all the lack of preclinical models that faithfully depict the local tumor microenvironment (TME), which is known to dramatically affect disease onset, progression and response to therapy. In this review, we provide the reader with a detailed overview of current 3D models developed to mimick the complexity and the dynamics of the TME, with a focus on understanding why the TME is a major target in anticancer therapy. We highlight the advantages and translational potentials of tumor spheroids, organoids and immune Tumor-on-a-Chip models in disease modeling and therapeutic response, while outlining pending challenges and limitations. Thinking forward, we focus on the possibility to integrate the know-hows of micro-engineers, cancer immunologists, pharmaceutical researchers and bioinformaticians to meet the needs of cancer researchers and clinicians interested in using these platforms with high fidelity for patient-tailored disease modeling and drug discovery.
Collapse
Affiliation(s)
- Nicoletta Manduca
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ester Maccafeo
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ruggero De Maria
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario ‘A. Gemelli’ - Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Antonella Sistigu
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Martina Musella
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
46
|
Alkim E, Dowst H, DiCarlo J, Dobrolecki LE, Hernández-Herrera A, Hormuth DA, Liao Y, McOwiti A, Pautler R, Rimawi M, Roark A, Srinivasan RR, Virostko J, Zhang B, Zheng F, Rubin DL, Yankeelov TE, Lewis MT. Toward Practical Integration of Omic and Imaging Data in Co-Clinical Trials. Tomography 2023; 9:810-828. [PMID: 37104137 PMCID: PMC10144684 DOI: 10.3390/tomography9020066] [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: 02/01/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/28/2023] Open
Abstract
Co-clinical trials are the concurrent or sequential evaluation of therapeutics in both patients clinically and patient-derived xenografts (PDX) pre-clinically, in a manner designed to match the pharmacokinetics and pharmacodynamics of the agent(s) used. The primary goal is to determine the degree to which PDX cohort responses recapitulate patient cohort responses at the phenotypic and molecular levels, such that pre-clinical and clinical trials can inform one another. A major issue is how to manage, integrate, and analyze the abundance of data generated across both spatial and temporal scales, as well as across species. To address this issue, we are developing MIRACCL (molecular and imaging response analysis of co-clinical trials), a web-based analytical tool. For prototyping, we simulated data for a co-clinical trial in "triple-negative" breast cancer (TNBC) by pairing pre- (T0) and on-treatment (T1) magnetic resonance imaging (MRI) from the I-SPY2 trial, as well as PDX-based T0 and T1 MRI. Baseline (T0) and on-treatment (T1) RNA expression data were also simulated for TNBC and PDX. Image features derived from both datasets were cross-referenced to omic data to evaluate MIRACCL functionality for correlating and displaying MRI-based changes in tumor size, vascularity, and cellularity with changes in mRNA expression as a function of treatment.
Collapse
Affiliation(s)
- Emel Alkim
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Heidi Dowst
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Julie DiCarlo
- Oden Institute for Computational Engineering and Sciences, Austin, TX 78712, USA
- Livestrong Cancer Institutes, Austin, TX 78712, USA
| | - Lacey E Dobrolecki
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology and Radiology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - David A Hormuth
- Oden Institute for Computational Engineering and Sciences, Austin, TX 78712, USA
- Livestrong Cancer Institutes, Austin, TX 78712, USA
| | - Yuxing Liao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Apollo McOwiti
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Robia Pautler
- Department of Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mothaffar Rimawi
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ashley Roark
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Jack Virostko
- Oden Institute for Computational Engineering and Sciences, Austin, TX 78712, USA
- Livestrong Cancer Institutes, Austin, TX 78712, USA
- Department of Oncology, The University of Texas at Austin, Austin, TX 78712, USA
- Department of Diagnostic Medicine, The University of Texas at Austin, Austin, TX 78712, USA
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Fei Zheng
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Daniel L Rubin
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Thomas E Yankeelov
- Oden Institute for Computational Engineering and Sciences, Austin, TX 78712, USA
- Livestrong Cancer Institutes, Austin, TX 78712, USA
- Department of Oncology, The University of Texas at Austin, Austin, TX 78712, USA
- Department of Diagnostic Medicine, The University of Texas at Austin, Austin, TX 78712, USA
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael T Lewis
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology and Radiology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
47
|
Balamurugan K, Poria DK, Sehareen SW, Krishnamurthy S, Tang W, McKennett L, Padmanaban V, Czarra K, Ewald AJ, Ueno NT, Ambs S, Sharan S, Sterneck E. Stabilization of E-cadherin adhesions by COX-2/GSK3β signaling is a targetable pathway in metastatic breast cancer. JCI Insight 2023; 8:156057. [PMID: 36757813 PMCID: PMC10070121 DOI: 10.1172/jci.insight.156057] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/07/2023] [Indexed: 02/10/2023] Open
Abstract
Metastatic progression of epithelial cancers can be associated with epithelial-mesenchymal transition (EMT) including transcriptional inhibition of E-cadherin (CDH1) expression. Recently, EM plasticity (EMP) and E-cadherin-mediated, cluster-based metastasis and treatment resistance have become more appreciated. However, the mechanisms that maintain E-cadherin expression in this context are less understood. Through studies of inflammatory breast cancer (IBC) and a 3D tumor cell "emboli" culture paradigm, we discovered that cyclooxygenase 2 (COX-2; PTGS2), a target gene of C/EBPδ (CEBPD), or its metabolite prostaglandin E2 (PGE2) promotes protein stability of E-cadherin, β-catenin, and p120 catenin through inhibition of GSK3β. The COX-2 inhibitor celecoxib downregulated E-cadherin complex proteins and caused cell death. Coexpression of E-cadherin and COX-2 was seen in breast cancer tissues from patients with poor outcome and, along with inhibitory GSK3β phosphorylation, in patient-derived xenografts (PDX) including triple negative breast cancer (TNBC).Celecoxib alone decreased E-cadherin protein expression within xenograft tumors, though CDH1 mRNA levels increased, and reduced circulating tumor cell (CTC) clusters. In combination with paclitaxel, celecoxib attenuated or regressed lung metastases. This study has uncovered a mechanism by which metastatic breast cancer cells can maintain E-cadherin-mediated cell-to-cell adhesions and cell survival, suggesting that some patients with COX-2+/E-cadherin+ breast cancer may benefit from targeting of the PGE2 signaling pathway.
Collapse
Affiliation(s)
- Kuppusamy Balamurugan
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, Maryland, USA
| | - Dipak K Poria
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, Maryland, USA
| | - Saadiya W Sehareen
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, Maryland, USA
| | - Savitri Krishnamurthy
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wei Tang
- Laboratory of Human Carcinogenesis, CCR, NCI, Bethesda, Maryland, USA
| | - Lois McKennett
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Veena Padmanaban
- Departments of Cell Biology and Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kelli Czarra
- Laboratory Animal Sciences Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Andrew J Ewald
- Departments of Cell Biology and Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Naoto T Ueno
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, CCR, NCI, Bethesda, Maryland, USA
| | - Shikha Sharan
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, Maryland, USA
| | - Esta Sterneck
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research (CCR), National Cancer Institute (NCI), Frederick, Maryland, USA
| |
Collapse
|
48
|
Peehl DM, Badea CT, Chenevert TL, Daldrup-Link HE, Ding L, Dobrolecki LE, Houghton AM, Kinahan PE, Kurhanewicz J, Lewis MT, Li S, Luker GD, Ma CX, Manning HC, Mowery YM, O’Dwyer PJ, Pautler RG, Rosen MA, Roudi R, Ross BD, Shoghi KI, Sriram R, Talpaz M, Wahl RL, Zhou R. Animal Models and Their Role in Imaging-Assisted Co-Clinical Trials. Tomography 2023; 9:657-680. [PMID: 36961012 PMCID: PMC10037611 DOI: 10.3390/tomography9020053] [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: 01/26/2023] [Revised: 03/08/2023] [Accepted: 03/08/2023] [Indexed: 03/19/2023] Open
Abstract
The availability of high-fidelity animal models for oncology research has grown enormously in recent years, enabling preclinical studies relevant to prevention, diagnosis, and treatment of cancer to be undertaken. This has led to increased opportunities to conduct co-clinical trials, which are studies on patients that are carried out parallel to or sequentially with animal models of cancer that mirror the biology of the patients' tumors. Patient-derived xenografts (PDX) and genetically engineered mouse models (GEMM) are considered to be the models that best represent human disease and have high translational value. Notably, one element of co-clinical trials that still needs significant optimization is quantitative imaging. The National Cancer Institute has organized a Co-Clinical Imaging Resource Program (CIRP) network to establish best practices for co-clinical imaging and to optimize translational quantitative imaging methodologies. This overview describes the ten co-clinical trials of investigators from eleven institutions who are currently supported by the CIRP initiative and are members of the Animal Models and Co-clinical Trials (AMCT) Working Group. Each team describes their corresponding clinical trial, type of cancer targeted, rationale for choice of animal models, therapy, and imaging modalities. The strengths and weaknesses of the co-clinical trial design and the challenges encountered are considered. The rich research resources generated by the members of the AMCT Working Group will benefit the broad research community and improve the quality and translational impact of imaging in co-clinical trials.
Collapse
Affiliation(s)
- Donna M. Peehl
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA; (J.K.); (R.S.)
| | - Cristian T. Badea
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA;
| | - Thomas L. Chenevert
- Department of Radiology and the Center for Molecular Imaging, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; (T.L.C.); (G.D.L.); (B.D.R.)
| | - Heike E. Daldrup-Link
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, CA 94305, USA; (H.E.D.-L.); (R.R.)
| | - Li Ding
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; (L.D.); (S.L.); (C.X.M.)
| | - Lacey E. Dobrolecki
- Advanced Technology Cores, Baylor College of Medicine, Houston, TX 77030, USA;
| | | | - Paul E. Kinahan
- Department of Radiology, University of Washington, Seattle, WA 98105, USA;
| | - John Kurhanewicz
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA; (J.K.); (R.S.)
| | - Michael T. Lewis
- Departments of Molecular and Cellular Biology and Radiology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Shunqiang Li
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; (L.D.); (S.L.); (C.X.M.)
| | - Gary D. Luker
- Department of Radiology and the Center for Molecular Imaging, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; (T.L.C.); (G.D.L.); (B.D.R.)
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Cynthia X. Ma
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; (L.D.); (S.L.); (C.X.M.)
| | - H. Charles Manning
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Yvonne M. Mowery
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27708, USA;
- Department of Head and Neck Surgery & Communication Sciences, Duke University School of Medicine, Durham, NC 27708, USA
| | - Peter J. O’Dwyer
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA; (P.J.O.); (M.A.R.); (R.Z.)
| | - Robia G. Pautler
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Mark A. Rosen
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA; (P.J.O.); (M.A.R.); (R.Z.)
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Raheleh Roudi
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Stanford University, Stanford, CA 94305, USA; (H.E.D.-L.); (R.R.)
| | - Brian D. Ross
- Department of Radiology and the Center for Molecular Imaging, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA; (T.L.C.); (G.D.L.); (B.D.R.)
- Department of Biological Chemistry, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Kooresh I. Shoghi
- Mallinckrodt Institute of Radiology (MIR), Washington University School of Medicine, St. Louis, MO 63110, USA; (K.I.S.); (R.L.W.)
| | - Renuka Sriram
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94158, USA; (J.K.); (R.S.)
| | - Moshe Talpaz
- Division of Hematology/Oncology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA;
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Richard L. Wahl
- Mallinckrodt Institute of Radiology (MIR), Washington University School of Medicine, St. Louis, MO 63110, USA; (K.I.S.); (R.L.W.)
| | - Rong Zhou
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA; (P.J.O.); (M.A.R.); (R.Z.)
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
49
|
Boyd DC, Zboril EK, Olex AL, Leftwich TJ, Hairr NS, Byers HA, Valentine AD, Altman JE, Alzubi MA, Grible JM, Turner SA, Ferreira-Gonzalez A, Dozmorov MG, Harrell JC. Discovering Synergistic Compounds with BYL-719 in PI3K Overactivated Basal-like PDXs. Cancers (Basel) 2023; 15:cancers15051582. [PMID: 36900375 PMCID: PMC10001201 DOI: 10.3390/cancers15051582] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/23/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Basal-like triple-negative breast cancer (TNBC) tumor cells are difficult to eliminate due to resistance mechanisms that promote survival. While this breast cancer subtype has low PIK3CA mutation rates when compared to estrogen receptor-positive (ER+) breast cancers, most basal-like TNBCs have an overactive PI3K pathway due to gene amplification or high gene expression. BYL-719 is a PIK3CA inhibitor that has been found to have low drug-drug interactions, which increases the likelihood that it could be useful for combinatorial therapy. Alpelisib (BYL-719) with fulvestrant was recently approved for treating ER+ breast cancer patients whose cancer had developed resistance to ER-targeting therapy. In these studies, a set of basal-like patient-derived xenograft (PDX) models was transcriptionally defined with bulk and single-cell RNA-sequencing and clinically actionable mutation profiles defined with Oncomine mutational profiling. This information was overlaid onto therapeutic drug screening results. BYL-719-based, synergistic two-drug combinations were identified with 20 different compounds, including everolimus, afatinib, and dronedarone, which were also found to be effective at minimizing tumor growth. These data support the use of these drug combinations towards cancers with activating PIK3CA mutations/gene amplifications or PTEN deficient/PI3K overactive pathways.
Collapse
Affiliation(s)
- David C. Boyd
- Department of Pathology, Virginia Commonwealth University, Richmond, VA 23298, USA
- Integrative Life Sciences Program, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Emily K. Zboril
- Department of Pathology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Amy L. Olex
- C. Kenneth and Dianne Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Tess J. Leftwich
- Department of Pathology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Nicole S. Hairr
- Department of Pathology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Holly A. Byers
- Department of Pathology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Aaron D. Valentine
- Department of Pathology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Julia E. Altman
- Department of Pathology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Mohammad A. Alzubi
- Department of Pathology, Virginia Commonwealth University, Richmond, VA 23298, USA
- Integrative Life Sciences Program, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Jacqueline M. Grible
- Department of Pathology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Scott A. Turner
- Department of Pathology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | | | - Mikhail G. Dozmorov
- Department of Pathology, Virginia Commonwealth University, Richmond, VA 23298, USA
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA 23219, USA
| | - J. Chuck Harrell
- Department of Pathology, Virginia Commonwealth University, Richmond, VA 23298, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
- Correspondence:
| |
Collapse
|
50
|
Szabo PM, Vajdi A, Kumar N, Tolstorukov MY, Chen BJ, Edwards R, Ligon KL, Chasalow SD, Chow KH, Shetty A, Bolisetty M, Holloway JL, Golhar R, Kidd BA, Hull PA, Houser J, Vlach L, Siemers NO, Saha S. Cancer-associated fibroblasts are the main contributors to epithelial-to-mesenchymal signatures in the tumor microenvironment. Sci Rep 2023; 13:3051. [PMID: 36810872 PMCID: PMC9944255 DOI: 10.1038/s41598-023-28480-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 01/19/2023] [Indexed: 02/24/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is associated with tumor initiation, metastasis, and drug resistance. However, the mechanisms underlying these associations are largely unknown. We studied several tumor types to identify the source of EMT gene expression signals and a potential mechanism of resistance to immuno-oncology treatment. Across tumor types, EMT-related gene expression was strongly associated with expression of stroma-related genes. Based on RNA sequencing of multiple patient-derived xenograft models, EMT-related gene expression was enriched in the stroma versus parenchyma. EMT-related markers were predominantly expressed by cancer-associated fibroblasts (CAFs), cells of mesenchymal origin which produce a variety of matrix proteins and growth factors. Scores derived from a 3-gene CAF transcriptional signature (COL1A1, COL1A2, COL3A1) were sufficient to reproduce association between EMT-related markers and disease prognosis. Our results suggest that CAFs are the primary source of EMT signaling and have potential roles as biomarkers and targets for immuno-oncology therapies.
Collapse
Affiliation(s)
- Peter M. Szabo
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Princeton, NJ USA ,grid.428458.70000 0004 1792 8104Present Address: Fate Therapeutics, San Diego, CA USA
| | - Amir Vajdi
- grid.65499.370000 0001 2106 9910Dana-Farber Cancer Institute, Boston, MA USA ,grid.417993.10000 0001 2260 0793Present Address: Merck & Co., Inc., Kenilworth, NJ USA
| | | | | | - Benjamin J. Chen
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Cambridge, MA USA
| | - Robin Edwards
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Princeton, NJ USA ,grid.428496.5Present Address: Daiichi Sankyo, Inc., Princeton, NJ USA
| | - Keith L. Ligon
- grid.65499.370000 0001 2106 9910Dana-Farber Cancer Institute, Boston, MA USA
| | - Scott D. Chasalow
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Princeton, NJ USA
| | - Kin-Hoe Chow
- grid.65499.370000 0001 2106 9910Dana-Farber Cancer Institute, Boston, MA USA
| | - Aniket Shetty
- grid.65499.370000 0001 2106 9910Dana-Farber Cancer Institute, Boston, MA USA
| | - Mohan Bolisetty
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Princeton, NJ USA
| | - James L. Holloway
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Seattle, WA USA
| | - Ryan Golhar
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Princeton, NJ USA
| | - Brian A. Kidd
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Redwood City, CA USA
| | | | - Jeff Houser
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Redwood City, CA USA
| | - Logan Vlach
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Redwood City, CA USA ,grid.152326.10000 0001 2264 7217Present Address: Vanderbilt University, Nashville, TN USA
| | - Nathan O. Siemers
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Princeton, NJ USA ,Present Address: Fiveprime Group, Monterey, CA USA
| | - Saurabh Saha
- grid.419971.30000 0004 0374 8313Bristol Myers Squibb, Princeton, NJ USA ,Present Address: Centessa Pharmaceuticals, Cambridge, MA USA
| |
Collapse
|