210 results on '"Alana L. Welm"'
Search Results
52. Data from Invasive Lobular Carcinoma Cell Lines Are Characterized by Unique Estrogen-Mediated Gene Expression Patterns and Altered Tamoxifen Response
- Author
-
Steffi Oesterreich, Alana L. Welm, David J. Dabbs, Nancy E. Davidson, Uma R. Chandran, Guoying Wang, Soumya Luthra, Amir Bahreini, Kristine L. Cooper, and Matthew J. Sikora
- Abstract
Invasive lobular carcinoma (ILC) is a histologic subtype of breast cancer that is frequently associated with favorable outcomes, as approximately 90% of ILC express the estrogen receptor (ER). However, recent retrospective analyses suggest that patients with ILC receiving adjuvant endocrine therapy may not benefit as much as patients with invasive ductal carcinoma. On the basis of these observations, we characterized ER function and endocrine response in ILC models. The ER-positive ILC cell lines MDA MB 134VI (MM134) and SUM44PE were used to examine the ER-regulated transcriptome via gene expression microarray analyses and ER ChIP-Seq, and to examine response to endocrine therapy. In parallel, estrogen response was assessed in vivo in the patient-derived ILC xenograft HCI-013. We identified 915 genes that were uniquely E2 regulated in ILC cell lines versus other breast cancer cell lines, and a subset of these genes were also E2 regulated in vivo in HCI-013. MM134 cells were de novo tamoxifen resistant and were induced to grow by 4-hydroxytamoxifen, as well as other antiestrogens, as partial agonists. Growth was accompanied by agonist activity of tamoxifen on ER-mediated gene expression. Though tamoxifen induced cell growth, MM134 cells required fibroblast growth factor receptor (FGFR)-1 signaling to maintain viability and were sensitive to combined endocrine therapy and FGFR1 inhibition. Our observation that ER drives a unique program of gene expression in ILC cells correlates with the ability of tamoxifen to induce growth in these cells. Targeting growth factors using FGFR1 inhibitors may block survival pathways required by ILC and reverse tamoxifen resistance. Cancer Res; 74(5); 1463–74. ©2014 AACR.
- Published
- 2023
53. Supplementary Table 1 from Invasive Lobular Carcinoma Cell Lines Are Characterized by Unique Estrogen-Mediated Gene Expression Patterns and Altered Tamoxifen Response
- Author
-
Steffi Oesterreich, Alana L. Welm, David J. Dabbs, Nancy E. Davidson, Uma R. Chandran, Guoying Wang, Soumya Luthra, Amir Bahreini, Kristine L. Cooper, and Matthew J. Sikora
- Abstract
XLSX file - 2042K, Lists for genes regulated by E2 in MM134 and SUM44 cells, and for categories of genes based on cell type specificity.
- Published
- 2023
54. Supplementary Figure 2 from Invasive Lobular Carcinoma Cell Lines Are Characterized by Unique Estrogen-Mediated Gene Expression Patterns and Altered Tamoxifen Response
- Author
-
Steffi Oesterreich, Alana L. Welm, David J. Dabbs, Nancy E. Davidson, Uma R. Chandran, Guoying Wang, Soumya Luthra, Amir Bahreini, Kristine L. Cooper, and Matthew J. Sikora
- Abstract
PDF file - 52K, Motif analysis of ER binding sites identified in MM134 by ChIP-seq.
- Published
- 2023
55. Supplementary Table 3 from Invasive Lobular Carcinoma Cell Lines Are Characterized by Unique Estrogen-Mediated Gene Expression Patterns and Altered Tamoxifen Response
- Author
-
Steffi Oesterreich, Alana L. Welm, David J. Dabbs, Nancy E. Davidson, Uma R. Chandran, Guoying Wang, Soumya Luthra, Amir Bahreini, Kristine L. Cooper, and Matthew J. Sikora
- Abstract
XLSX file - 17K, Expression data from estrogen deprivation studies using PDX HCI-013.
- Published
- 2023
56. Transferred mitochondria accumulate reactive oxygen species, promoting proliferation
- Author
-
Joseph R Casalini, Chelsea U Kidwell, Soorya Pradeep, Sandra D Scherer, Daniel Greiner, Defne Bayik, Dionysios C Watson, Gregory S Olson, Justin D Lathia, Jarrod S Johnson, Jared Rutter, Alana L Welm, Thomas A Zangle, and Minna Roh-Johnson
- Subjects
General Immunology and Microbiology ,General Neuroscience ,General Medicine ,General Biochemistry, Genetics and Molecular Biology - Abstract
Recent studies reveal that lateral mitochondrial transfer, the movement of mitochondria from one cell to another, can affect cellular and tissue homeostasis. Most of what we know about mitochondrial transfer stems from bulk cell studies and have led to the paradigm that functional transferred mitochondria restore bioenergetics and revitalize cellular functions to recipient cells with damaged or non-functional mitochondrial networks. However, we show that mitochondrial transfer also occurs between cells with functioning endogenous mitochondrial networks, but the mechanisms underlying how transferred mitochondria can promote such sustained behavioral reprogramming remain unclear. We report that unexpectedly, transferred macrophage mitochondria are dysfunctional and accumulate reactive oxygen species in recipient cancer cells. We further discovered that reactive oxygen species accumulation activates ERK signaling, promoting cancer cell proliferation. Pro-tumorigenic macrophages exhibit fragmented mitochondrial networks, leading to higher rates of mitochondrial transfer to cancer cells. Finally, we observe that macrophage mitochondrial transfer promotes tumor cell proliferation in vivo. Collectively these results indicate that transferred macrophage mitochondria activate downstream signaling pathways in a ROS-dependent manner in cancer cells, and provide a model of how sustained behavioral reprogramming can be mediated by a relatively small amount of transferred mitochondria in vitro and in vivo.
- Published
- 2023
57. Low-affinity CAR T cells exhibit reduced trogocytosis, preventing rapid antigen loss, and increasing CAR T cell expansion
- Author
-
Michael L. Olson, Erica R. Vander Mause, Sabarinath V. Radhakrishnan, Joshua D. Brody, Aaron P. Rapoport, Alana L. Welm, Djordje Atanackovic, and Tim Luetkens
- Subjects
Cancer Research ,Oncology ,Hematology - Published
- 2022
58. A human breast cancer-derived xenograft and organoid platform for drug discovery and precision oncology
- Author
-
Katrin P. Guillen, Maihi Fujita, Andrew J. Butterfield, Sandra D. Scherer, Matthew H. Bailey, Zhengtao Chu, Yoko S. DeRose, Ling Zhao, Emilio Cortes-Sanchez, Chieh-Hsiang Yang, Jennifer Toner, Guoying Wang, Yi Qiao, Xiaomeng Huang, Jeffery A. Greenland, Jeffery M. Vahrenkamp, David H. Lum, Rachel E. Factor, Edward W. Nelson, Cindy B. Matsen, Jane M. Poretta, Regina Rosenthal, Anna C. Beck, Saundra S. Buys, Christos Vaklavas, John H. Ward, Randy L. Jensen, Kevin B. Jones, Zheqi Li, Steffi Oesterreich, Lacey E. Dobrolecki, Satya S. Pathi, Xing Yi Woo, Kristofer C. Berrett, Mark E. Wadsworth, Jeffrey H. Chuang, Michael T. Lewis, Gabor T. Marth, Jason Gertz, Katherine E. Varley, Bryan E. Welm, and Alana L. Welm
- Subjects
Organoids ,Cancer Research ,Oncology ,Drug Discovery ,Heterografts ,Humans ,Triple Negative Breast Neoplasms ,Precision Medicine ,Xenograft Model Antitumor Assays ,United States - Abstract
Models that recapitulate the complexity of human tumors are urgently needed to develop more effective cancer therapies. We report a bank of human patient-derived xenografts (PDXs) and matched organoid cultures from tumors that represent the greatest unmet need: endocrine-resistant, treatment-refractory and metastatic breast cancers. We leverage matched PDXs and PDX-derived organoids (PDxO) for drug screening that is feasible and cost-effective with in vivo validation. Moreover, we demonstrate the feasibility of using these models for precision oncology in real time with clinical care in a case of triple-negative breast cancer (TNBC) with early metastatic recurrence. Our results uncovered a Food and Drug Administration (FDA)-approved drug with high efficacy against the models. Treatment with this therapy resulted in a complete response for the individual and a progression-free survival (PFS) period more than three times longer than their previous therapies. This work provides valuable methods and resources for functional precision medicine and drug development for human breast cancer.
- Published
- 2022
59. Author response: Transferred mitochondria accumulate reactive oxygen species, promoting proliferation
- Author
-
Joseph R Casalini, Chelsea U Kidwell, Soorya Pradeep, Sandra D Scherer, Daniel Greiner, Defne Bayik, Dionysios C Watson, Gregory S Olson, Justin D Lathia, Jarrod S Johnson, Jared Rutter, Alana L Welm, Thomas A Zangle, and Minna Roh-Johnson
- Published
- 2023
60. Astrocytic laminin-211 drives disseminated breast tumor cell dormancy in brain
- Author
-
Jinxiang Dai, Patrick J. Cimino, Kenneth H. Gouin, Candice A. Grzelak, Alexander Barrett, Andrea R. Lim, Annalyssa Long, Stephanie Weaver, Lindsey T. Saldin, Aiyedun Uzamere, Vera Schulte, Nigel Clegg, Laura Pisarsky, David Lyden, Mina J. Bissell, Simon Knott, Alana L. Welm, Jason H. Bielas, Kirk C. Hansen, Frank Winkler, Eric C. Holland, and Cyrus M. Ghajar
- Subjects
Cancer Research ,Oncology ,Brain Neoplasms ,Astrocytes ,Tumor Microenvironment ,Brain ,Humans ,Breast Neoplasms ,Female ,Laminin - Abstract
Although dormancy is thought to play a key role in the metastasis of breast tumor cells to the brain, our knowledge of the molecular mechanisms regulating disseminated tumor cell (DTC) dormancy in this organ is limited. Here using serial intravital imaging of dormant and metastatic triple-negative breast cancer lines, we identify escape from the single-cell or micrometastatic state as the rate-limiting step towards brain metastasis. We show that every DTC occupies a vascular niche, with quiescent DTCs residing on astrocyte endfeet. At these sites, astrocyte-deposited laminin-211 drives DTC quiescence by inducing the dystroglycan receptor to associate with yes-associated protein, thereby sequestering it from the nucleus and preventing its prometastatic functions. These findings identify a brain-specific mechanism of DTC dormancy and highlight the need for a more thorough understanding of tumor dormancy to develop therapeutic approaches that prevent brain metastasis.
- Published
- 2021
61. Transcriptional Reprogramming Differentiates Active from Inactive ESR1 Fusions in Endocrine Therapy-Refractory Metastatic Breast Cancer
- Author
-
Dan R. Robinson, Viktoriya Korchina, Xuxu Gou, Nicholas Mitsiades, Shunqiang Li, Jianhong Hu, Harshavardhan Doddapaneni, Matthew J. Ellis, Purba Singh, Beom-Jun Kim, Alana L. Welm, Michael T. Lewis, Diana Fandino, Lacey E. Dobrolecki, Meenakshi Anurag, Sinem Seker, Jonathan T. Lei, Airi Han, Saif Rehman, Adrian V. Lee, and Charles E. Foulds
- Subjects
Cancer Research ,Antineoplastic Agents, Hormonal ,Oncogene Proteins, Fusion ,Somatic cell ,Mice, Nude ,Apoptosis ,Breast Neoplasms ,Biology ,Article ,Mice ,Biomarkers, Tumor ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Gene ,Cell Proliferation ,Fulvestrant ,Point mutation ,Estrogen Receptor alpha ,RNA ,Prognosis ,Xenograft Model Antitumor Assays ,Fusion protein ,Gene Expression Regulation, Neoplastic ,Survival Rate ,body regions ,Oncology ,Drug Resistance, Neoplasm ,Mutation ,Cancer research ,Female ,Transcriptome ,Reprogramming ,Estrogen receptor alpha ,medicine.drug - Abstract
Genomic analysis has recently identified multiple ESR1 gene translocations in estrogen receptor alpha–positive (ERα+) metastatic breast cancer (MBC) that encode chimeric proteins whereby the ESR1 ligand binding domain (LBD) is replaced by C-terminal sequences from many different gene partners. Here we functionally screened 15 ESR1 fusions and identified 10 that promoted estradiol-independent cell growth, motility, invasion, epithelial-to-mesenchymal transition, and resistance to fulvestrant. RNA sequencing identified a gene expression pattern specific to functionally active ESR1 gene fusions that was subsequently reduced to a diagnostic 24-gene signature. This signature was further examined in 20 ERα+ patient-derived xenografts and in 55 ERα+ MBC samples. The 24-gene signature successfully identified cases harboring ESR1 gene fusions and also accurately diagnosed the presence of activating ESR1 LBD point mutations. Therefore, the 24-gene signature represents an efficient approach to screening samples for the presence of diverse somatic ESR1 mutations and translocations that drive endocrine treatment failure in MBC. Significance: This study identifies a gene signature diagnostic for functional ESR1 fusions that drive poor outcome in advanced breast cancer, which could also help guide precision medicine approaches in patients harboring ESR1 mutations.
- Published
- 2021
62. A path to translation: How 3D patient tumor avatars enable next generation precision oncology
- Author
-
Margarida Barroso, Milan G. Chheda, Hans Clevers, Elena Elez, Salma Kaochar, Scott E. Kopetz, Xiao-Nan Li, Funda Meric-Bernstam, Clifford A. Meyer, Haiwei Mou, Kristen M. Naegle, Martin F. Pera, Zinaida Perova, Katerina A. Politi, Benjamin J. Raphael, Paul Robson, Rosalie C. Sears, Josep Tabernero, David A. Tuveson, Alana L. Welm, Bryan E. Welm, Christopher D. Willey, Konstantin Salnikow, Jeffrey H. Chuang, Xiling Shen, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
Cancer Research ,Oncology ,Neoplasms ,Humans ,Prospective Studies ,Precision Medicine ,Medical Oncology - Abstract
3D patient tumor avatars (3D-PTAs) hold promise for next-generation precision medicine. Here, we describe the benefits and challenges of 3D-PTA technologies and necessary future steps to realize their potential for clinical decision making. 3D-PTAs require standardization criteria and prospective trials to establish clinical benefits. Innovative trial designs that combine omics and 3D-PTA readouts may lead to more accurate clinical predictors, and an integrated platform that combines diagnostic and therapeutic development will accelerate new treatments for patients with refractory disease.
- Published
- 2022
63. Estrogen receptor alpha mutations regulate gene expression and cell growth in breast cancer through microRNAs
- Author
-
Spencer Arnesen, Jacob T. Polaski, Zannel Blanchard, Kyle S. Osborne, Alana L. Welm, Ryan M. O’Connell, and Jason Gertz
- Abstract
Estrogen receptor α (ER) mutations occur in up to 30% of metastatic ER-positive breast cancers. Recent data has shown that ER mutations impact the expression of thousands of genes not typically regulated by wildtype ER. While the majority of these altered genes can be explained by constant activity of mutant ER or genomic changes such as altered ER binding and chromatin accessibility, as much as 33% remain unexplained, indicating the potential for post-transcriptional effects. Here we explored the role of microRNAs in mutant ER-driven gene regulation and identified several microRNAs that are dysregulated in ER mutant cells. These differentially regulated microRNAs target a significant portion of mutant-specific genes involved in key cellular processes. When the activity of microRNAs is altered using mimics or inhibitors, significant changes are observed in gene expression and cellular proliferation related to mutant ER. An in-depth evaluation of miR-301b led us to discover an important role forPRKD3in the proliferation of ER mutant cells. Our findings show that microRNAs contribute to mutant ER gene regulation and cellular effects in breast cancer cells.
- Published
- 2022
64. Comprehensive characterization of 536 patient-derived xenograft models prioritizes candidatesfor targeted treatment
- Author
-
Li Chen, Jacqueline Mudd, Michael Ittmann, Carol J. Bult, Amanda R. Kirane, Jelena Randjelovic, Stephen Scott, Yige Wu, Li Ding, Vashisht G. Yennu-Nanda, Jing Wang, Christopher D. Lanier, Maihi Fujita, Emilio Cortes-Sanchez, Sienna Rocha, Susan G. Hilsenbeck, Kian-Huat Lim, Fernanda Martins Rodrigues, Jill Rubinstein, Nicholas Mitsiades, Haiyin Lin, Jayamanna Wickramasinghe, Andrew Butterfield, Bryan E. Welm, Alana L. Welm, Jose P. Zevallos, Jason Held, Nicole B. Coggins, Song Cao, Yuanxin Xi, Brenda C. Timmons, Paul Lott, David Menter, Shunqiang Li, Tina Primeau, Fei Yang, Andrea Wang-Gillam, Ramaswamy Govindan, Dali Li, Brandi Davis-Dusenbery, Sara Seepo, Michael C. Wendl, Jeffrey Grover, Brian S. White, Clifford G. Tepper, Peter N. Robinson, Michael A. Davies, Zhengtao Chu, Michael W. Lloyd, Hua Sun, Xiaoshan Zhang, Tamara Stankovic, Dylan Fingerman, Anuj Srivastava, Luis G. Carvajal-Carmona, Don L. Gibbons, Lijun Yao, Rebecca Aft, Hongyong Zhang, Ismail Meraz, John DiGiovanna, Scott Kopetz, Ling Zhao, Guadalupe Polanco-Echeverry, Feng Chen, Jeremy Hoog, Matthew A. Wyczalkowski, George Xu, John D. Minna, Yi Xu, Julie Belmar, Xiaowei Xu, Luc Girard, Dennis A. Dean, Tijana Borovski, Chong-xian Pan, Cynthia X. Ma, Alexa Morales Arana, Yize Li, Turcin Saridogan, Steven B. Neuhauser, Sandra Scherer, Vicki Chin, Rose Tipton, David R. Gandara, Sherri R. Davies, Argun Akcakanat, Rajesh Patidar, Julie K. Schwarz, Soner Koc, Gao Boning, Michael Kim, Bryce P. Kirby, Yvonne A. Evrard, Hyunsil Park, Christian Frech, Chia-Kuei Mo, Ran Zhang, Brian A. Van Tine, Jonathan W. Reiss, Min Xiao, Xing Yi Woo, Tiffany Le, Ana Estrada, Xiaofeng Zheng, Jeffrey A. Moscow, Mourad Majidi, Nadezhda V. Terekhanova, Katherine Fuh, Erkan Yuca, Timothy A. Yap, Jianhua Zhang, Matthew J. Ellis, Shannon Westin, James H. Doroshow, Vito W. Rebecca, Moon S. Chen, Coya Tapia, Reyka G Jayasinghe, Jack A. Roth, Jithesh Augustine, Ryan C. Fields, Michae T. Tetzlaff, Michael T. Lewis, Kurt W. Evans, Ralph W. deVere White, Brian J. Sanderson, May Cho, Jeffrey H. Chuang, Tiffany Wallace, Ryan Jeon, Ted Toal, Matthew H. Bailey, Bert W. O'Malley, Katherine L. Nathanson, Qin Liu, Benjamin J. Raphael, Jingqin Luo, Salma Kaochar, Huiqin Chen, Rajasekharan Somasundaram, Daniel Cui Zhou, John F. DiPersio, Andrew V. Kossenkov, Bingliang Fang, Vanessa Jensen, Simone Zaccaria, Alexey Sorokin, Ai-Hong Ma, Sidharth V. Puram, Min Jin Ha, Meenhard Herlyn, R. Jay Mashl, Kelly Gale, Bingbing Dai, Lacey E. Dobrolecki, Chieh-Hsiang Yang, and Funda Meric-Bernstam
- Subjects
endocrine system ,Science ,Druggability ,General Physics and Astronomy ,Genomics ,Computational biology ,Biology ,Genome ,digestive system ,General Biochemistry, Genetics and Molecular Biology ,Article ,Research community ,Multiple time ,medicine ,Cancer genomics ,Cancer models ,Tumor xenograft ,Multidisciplinary ,Cancer ,General Chemistry ,medicine.disease ,Pharmacogenomics ,Data integration ,hormones, hormone substitutes, and hormone antagonists - Abstract
Development of candidate cancer treatments is a resource-intensive process, with the research community continuing to investigate options beyond static genomic characterization. Toward this goal, we have established the genomic landscapes of 536 patient-derived xenograft (PDX) models across 25 cancer types, together with mutation, copy number, fusion, transcriptomic profiles, and NCI-MATCH arms. Compared with human tumors, PDXs typically have higher purity and fit to investigate dynamic driver events and molecular properties via multiple time points from same case PDXs. Here, we report on dynamic genomic landscapes and pharmacogenomic associations, including associations between activating oncogenic events and drugs, correlations between whole-genome duplications and subclone events, and the potential PDX models for NCI-MATCH trials. Lastly, we provide a web portal having comprehensive pan-cancer PDX genomic profiles and source code to facilitate identification of more druggable events and further insights into PDXs’ recapitulation of human tumors., Patient-derived xenograft models (PDX) have been extensively used to study the molecular and clinical features of cancers. Here the authors present a cohort of 536 PDX models from 25 cancers, as well as their genomic and evolutionary profiles and their suitability for clinical trials.
- Published
- 2021
65. Breast Cancer Immunity: It is TIME for the Next Chapter
- Author
-
Daniela F. Quail, Morag Park, Alana L. Welm, and H. Atakan Ekiz
- Subjects
General Biochemistry, Genetics and Molecular Biology - Published
- 2023
66. Abstract 5407: A pan-cancer PDX histology image repository with genomic and pathological annotations for deep learning analysis
- Author
-
Brian S. White, Xing Yi Woo, Soner Koc, Todd Sheridan, Steven B. Neuhauser, Shidan Wang, Yvonne A. Evrard, John David Landua, R Jay Mashl, Sherri R. Davies, Bingliang Fang, Maria Gabriela Raso, Kurt W. Evans, Matthew H. Bailey, Yeqing Chen, Min Xiao, Jill Rubinstein, Ali Foroughi pour, Lacey Elizabeth Dobrolecki, Maihi Fujita, Junya Fujimoto, Guanghua Xiao, Ryan C. Fields, Jacqueline L. Mudd, Xiaowei Xu, Melinda G. Hollingshead, Shahanawaz Jiwani, PDXNet consortium, Tiffany A. Wallace, Jeffrey A. Moscow, James H. Doroshow, Nicholas Mitsiades, Salma Kaochar, Chong-xian Pan, Moon S. Chen, Luis G. Carvajal-Carmona, Alana L. Welm, Bryan E. Welm, Ramaswamy Govindan, Shunqiang Li, Michael A. Davies, Jack A. Roth, Funda Meric-Bernstam, Yang Xie, Meenhard Herlyn, Li Ding, Michael T. Lewis, Carol J. Bolt, Dennis A. Dean, and Jeffrey H. Chuang
- Subjects
Cancer Research ,Oncology - Abstract
Patient-derived xenografts (PDXs) model human intra-tumoral heterogeneity in the context of the intact tissue of immunocompromised mice. Histological imaging via hematoxylin and eosin (H&E) staining is performed on PDX samples for routine assessment and, in principle, captures the complex interplay between tumor and stromal cells. Deep learning (DL)-based analysis of large human H&E image repositories has extracted inter-cellular and morphological signals correlated with disease phenotype and therapeutic response. Here, we present an extensive, pan-cancer repository of nearly 1,000 PDX and paired human progenitor H&E images. These images, curated from the PDXNet consortium, are associated with genomic and transcriptomic data, clinical metadata, pathological assessment of cell composition, and, in several cases, detailed pathological annotation of tumor, stroma, and necrotic regions. We demonstrate that DL can be applied to these images to classify tumor regions with an accuracy of 0.87. Further, we show that DL can predict xenograft-transplant lymphoproliferative disorder, the unintended outgrowth of human lymphocytes at the transplantation site, with an accuracy of 0.97. This repository enables PDX-specific investigations of cancer biology through histopathological analysis and contributes important model system data that expand on existing human histology repositories. We expect the PDXNet Image Repository to be valuable for controlled digital pathology analysis, both for the evaluation of technical issues such as stain normalization and for development of novel computational methods based on spatial behaviors within cancer tissues. Citation Format: Brian S. White, Xing Yi Woo, Soner Koc, Todd Sheridan, Steven B. Neuhauser, Shidan Wang, Yvonne A. Evrard, John David Landua, R Jay Mashl, Sherri R. Davies, Bingliang Fang, Maria Gabriela Raso, Kurt W. Evans, Matthew H. Bailey, Yeqing Chen, Min Xiao, Jill Rubinstein, Ali Foroughi pour, Lacey Elizabeth Dobrolecki, Maihi Fujita, Junya Fujimoto, Guanghua Xiao, Ryan C. Fields, Jacqueline L. Mudd, Xiaowei Xu, Melinda G. Hollingshead, Shahanawaz Jiwani, PDXNet consortium, Tiffany A. Wallace, Jeffrey A. Moscow, James H. Doroshow, Nicholas Mitsiades, Salma Kaochar, Chong-xian Pan, Moon S. Chen, Luis G. Carvajal-Carmona, Alana L. Welm, Bryan E. Welm, Ramaswamy Govindan, Shunqiang Li, Michael A. Davies, Jack A. Roth, Funda Meric-Bernstam, Yang Xie, Meenhard Herlyn, Li Ding, Michael T. Lewis, Carol J. Bolt, Dennis A. Dean, Jeffrey H. Chuang. A pan-cancer PDX histology image repository with genomic and pathological annotations for deep learning analysis. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 5407.
- Published
- 2023
67. Abstract PS17-24: Inhibition of short-form ron eliminates breast cancer metastases through an immune-mediated mechanism
- Author
-
Huseyin Atakan Ekiz, Alana L. Welm, Harika Gundlapalli, and Shu-Chin Alicia Lai
- Subjects
Cancer Research ,biology ,business.industry ,medicine.medical_treatment ,Cancer ,Immunosuppression ,Immunotherapy ,medicine.disease ,Metastatic breast cancer ,Receptor tyrosine kinase ,Metastasis ,Breast cancer ,Immune system ,Oncology ,medicine ,Cancer research ,biology.protein ,business - Abstract
Metastatic breast cancer is the overwhelming cause of breast cancer mortality and is still incurable. The rapid development of immunotherapy is an exciting new area of research in metastatic breast cancer. However, the extreme immunosuppressive tumor environment poses a major challenge. A better understanding of how the immune system can be harnessed against metastatic cancer is required to improve patient outcomes. We previously showed that expression of the receptor tyrosine kinase Ron in the host, rather than Ron's tumor expression, contributed to tumor-associated immunosuppression and duo inhibition of Ron and CTLA-4 significantly reduced metastatic outgrowth. However, the actual mechanism remains unclear. The present study provides evidence that the N-terminal truncated isoform, short-form Ron (SF-Ron), is the major contributor in suppressing the anti-tumor immune responses and promoting metastatic outgrowth. Genetic deletion of host SF-Ron nearly eliminated breast cancer metastasis in mice, lead to systemic immune-activation, increased recruitment of lymphocytes to the site of metastasis, and augmented tumor-specific T-cell responses. Lack of SF-Ron also leads to the accumulation of CD4+ T-cells in the metastatic lungs and endowed with anti-tumor potential. Importantly, mice treated with small molecule Ron kinase inhibitor that targets both Ron and SF-Ron, produced significantly higher, active, tumor-specific CD8+ T-cells. Our study indicates that blocking Ron, especially the SF-Ron, remodels the metastatic lung microenvironment to enhance anti-tumor immunity. This study sheds light on the potential non-redundant roles of full-length and SF-Ron isoforms in mediating breast cancer metastasis and anti-tumor immune responses; and highlights the relevance of combining Ron inhibitors with immunotherapeutic agents to potentially improve treatment efficacy for metastatic breast cancer patients. Citation Format: Shu-Chin Alicia Lai, Harika Gundlapalli, Huseyin A Ekiz, Alana L Welm. Inhibition of short-form ron eliminates breast cancer metastases through an immune-mediated mechanism [abstract]. In: Proceedings of the 2020 San Antonio Breast Cancer Virtual Symposium; 2020 Dec 8-11; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2021;81(4 Suppl):Abstract nr PS17-24.
- Published
- 2021
68. Dll1+ quiescent tumor stem cells drive chemoresistance in breast cancer through NF-κB survival pathway
- Author
-
John W. Tobias, Mario Andres Blanco, Snahlata Singh, Sushil Kumar, Alana L. Welm, Christopher J. Lengner, Rumela Chakrabarti, Yibin Kang, Christian W. Siebel, Andres J. Klein-Szanto, Ajeya Nandi, Rohan Regulapati, and Ning Li
- Subjects
0301 basic medicine ,medicine.medical_treatment ,General Physics and Astronomy ,Datasets as Topic ,Metastasis ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Breast cancer ,Conditional gene knockout ,Antineoplastic Combined Chemotherapy Protocols ,Breast ,RNA-Seq ,Mice, Knockout ,Multidisciplinary ,Receptors, Notch ,Cancer stem cells ,Phenotype ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,Benzamides ,Neoplastic Stem Cells ,Female ,Signal Transduction ,Cell Survival ,Science ,Breast Neoplasms ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Cell Proliferation ,Chemotherapy ,business.industry ,Calcium-Binding Proteins ,Cancer ,Membrane Proteins ,NF-kappa B p50 Subunit ,NF-κB ,General Chemistry ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,chemistry ,Doxorubicin ,Drug Resistance, Neoplasm ,Cancer cell ,Cancer research ,business - Abstract
Development of chemoresistance in breast cancer patients greatly increases mortality. Thus, understanding mechanisms underlying breast cancer resistance to chemotherapy is of paramount importance to overcome this clinical challenge. Although activated Notch receptors have been associated with chemoresistance in cancer, the specific Notch ligands and their molecular mechanisms leading to chemoresistance in breast cancer remain elusive. Using conditional knockout and reporter mouse models, we demonstrate that tumor cells expressing the Notch ligand Dll1 is important for tumor growth and metastasis and bear similarities to tumor-initiating cancer cells (TICs) in breast cancer. RNA-seq and ATAC-seq using reporter models and patient data demonstrated that NF-κB activation is downstream of Dll1 and is associated with a chemoresistant phenotype. Finally, pharmacological blocking of Dll1 or NF-κB pathway completely sensitizes Dll1+ tumors to chemotherapy, highlighting therapeutic avenues for chemotherapy resistant breast cancer patients in the near future., Although activated Notch receptors have been associated with chemoresistance in cancer, the role of specific Notch ligands remain elusive. Here, the authors show that in breast cells the Notch ligand DLL1 is expressed in cells with a cancer stem cell phenotype and promote doxorubicin resistance in part through NF-kB, as well as metastasis.
- Published
- 2021
69. RON signalling promotes therapeutic resistance in ESR1 mutant breast cancer
- Author
-
Jean Paul De La O, Hangqing Lin, Shunqiang Li, Beom-Jun Kim, Alana L. Welm, David Edwards, Doug W. Chan, Sandra L. Grimm, Guowei Gu, Dan Liu, Derek Dustin, Amanda Beyer, Suzanne A. W. Fuqua, Thomas Gonzalez, Cristian Coarfa, Sarah Herzog, and Matthew J. Ellis
- Subjects
Cancer Research ,Pyridines ,Mutant ,Breast Neoplasms ,Piperazines ,Article ,Metastasis ,Mice ,Breast cancer ,medicine ,Animals ,Humans ,Kinome ,Aromatase ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,biology ,Kinase ,business.industry ,Estrogen Receptor alpha ,Receptor Protein-Tyrosine Kinases ,medicine.disease ,Xenograft Model Antitumor Assays ,Metastatic breast cancer ,body regions ,Oncology ,Drug Resistance, Neoplasm ,Mutation ,Cancer research ,biology.protein ,Female ,business ,Estrogen receptor alpha ,Signal Transduction - Abstract
Background Oestrogen Receptor 1 (ESR1) mutations are frequently acquired in oestrogen receptor (ER)-positive metastatic breast cancer (MBC) patients who were treated with aromatase inhibitors (AI) in the metastatic setting. Acquired ESR1 mutations are associated with poor prognosis and there is a lack of effective therapies that selectively target these cancers. Methods We performed a proteomic kinome analysis in ESR1 Y537S mutant cells to identify hyperactivated kinases in ESR1 mutant cells. We validated Recepteur d’Origine Nantais (RON) and PI3K hyperactivity through phospho-immunoblot analysis, organoid growth assays, and in an in vivo patient-derived xenograft (PDX) metastatic model. Results We demonstrated that RON was hyperactivated in ESR1 mutant models, and in acquired palbociclib-resistant (PalbR) models. RON and insulin-like growth factor 1 receptor (IGF-1R) interacted as shown through pharmacological and genetic inhibition and were regulated by the mutant ER as demonstrated by reduced phospho-protein expression with endocrine therapies (ET). We show that ET in combination with a RON inhibitor (RONi) decreased ex vivo organoid growth of ESR1 mutant models, and as a monotherapy in PalbR models, demonstrating its therapeutic efficacy. Significantly, ET in combination with the RONi reduced metastasis of an ESR1 Y537S mutant PDX model. Conclusions Our results demonstrate that RON/PI3K pathway inhibition may be an effective treatment strategy in ESR1 mutant and PalbR MBC patients. Clinically our data predict that ET resistance mechanisms can also contribute to CDK4/6 inhibitor resistance.
- Published
- 2020
70. PDXNet portal: patient-derived Xenograft model, data, workflow and tool discovery
- Author
-
Soner, Koc, Michael W, Lloyd, Jeffrey W, Grover, Nan, Xiao, Sara, Seepo, Sai Lakshmi, Subramanian, Manisha, Ray, Christian, Frech, John, DiGiovanna, Phillip, Webster, Steven, Neuhauser, Anuj, Srivastava, Xing Yi, Woo, Brian J, Sanderson, Brian, White, Paul, Lott, Lacey E, Dobrolecki, Heidi, Dowst, Yvonne A, Evrard, Tiffany A, Wallace, Jeffrey A, Moscow, James H, Doroshow, Nicholas, Mitsiades, Salma, Kaochar, Chong-Xian, Pan, Moon S, Chen, Luis, Carvajal-Carmona, Alana L, Welm, Bryan E, Welm, Michael T, Lewis, Ramaswamy, Govindan, Li, Ding, Shunqiang, Li, Meenhard, Herlyn, Michael A, Davies, Jack, Roth, Funda, Meric-Bernstam, Peter N, Robinson, Carol J, Bult, Brandi, Davis-Dusenbery, Dennis A, Dean, and Ai-Hong, Ma
- Subjects
General Medicine - Abstract
We created the PDX Network (PDXNet) portal (https://portal.pdxnetwork.org/) to centralize access to the National Cancer Institute-funded PDXNet consortium resources, to facilitate collaboration among researchers and to make these data easily available for research. The portal includes sections for resources, analysis results, metrics for PDXNet activities, data processing protocols and training materials for processing PDX data. Currently, the portal contains PDXNet model information and data resources from 334 new models across 33 cancer types. Tissue samples of these models were deposited in the NCI’s Patient-Derived Model Repository (PDMR) for public access. These models have 2134 associated sequencing files from 873 samples across 308 patients, which are hosted on the Cancer Genomics Cloud powered by Seven Bridges and the NCI Cancer Data Service for long-term storage and access with dbGaP permissions. The portal includes results from freely available, robust, validated and standardized analysis workflows on PDXNet sequencing files and PDMR data (3857 samples from 629 patients across 85 disease types). The PDXNet portal is continuously updated with new data and is of significant utility to the cancer research community as it provides a centralized location for PDXNet resources, which support multi-agent treatment studies, determination of sensitivity and resistance mechanisms, and preclinical trials.
- Published
- 2022
71. The lingering mysteries of metastatic recurrence in breast cancer
- Author
-
Katherine E. Varley, Alana L. Welm, and Alessandra I. Riggio
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Breast Neoplasms ,Review Article ,Disease ,Systemic therapy ,Metastasis ,03 medical and health sciences ,Breast cancer ,0302 clinical medicine ,Neoplasm Recurrence ,Internal medicine ,Animals ,Humans ,Medicine ,Neoplasm Invasiveness ,Neoplasm Metastasis ,030304 developmental biology ,0303 health sciences ,business.industry ,Extramural ,Cancer ,medicine.disease ,Key factors ,030220 oncology & carcinogenesis ,Female ,Neoplasm Recurrence, Local ,business - Abstract
Despite being the hallmark of cancer that is responsible for the highest number of deaths, very little is known about the biology of metastasis. Metastatic disease typically manifests after a protracted period of undetectable disease following surgery or systemic therapy, owing to relapse or recurrence. In the case of breast cancer, metastatic relapse can occur months to decades after initial diagnosis and treatment. In this review, we provide an overview of the known key factors that influence metastatic recurrence, with the goal of highlighting the critical unanswered questions that still need to be addressed to make a difference in the mortality of breast cancer patients.
- Published
- 2020
72. Enrichment of Collagen Fragments Using Dimeric Collagen Hybridizing Peptide for Urinary Collagenomics
- Author
-
S. Michael Yu, Julian L. Kessler, Alana L. Welm, Yang Li, and Jaime Fornetti
- Subjects
chemistry.chemical_classification ,Disease detection ,Chemistry ,Urinary system ,Peptide ,General Chemistry ,Biochemistry ,Article ,Peptide Fragments ,Biological fluid ,Extracellular matrix ,Mice ,Tandem Mass Spectrometry ,Liquid chromatography–mass spectrometry ,Animals ,Collagen ,Swissprot database ,Biomarker discovery ,Peptides ,Chromatography, Liquid - Abstract
Collagen remodeling in normal and pathologic conditions releases numerous collagen fragments into biological fluids. Although a few collagen fragments have been tested as biomarkers for disease indication, most occur at trace levels, making them nearly impossible to detect even with modern analytical tools. Here we report a new way to enrich collagen fragments that allows complete peptidomic analysis of collagen fragments in urine. Enrichment is made possible by dimeric collagen hybridizing peptides (CHPs) that bind collagen fragments originating from the triple helical regions of all collagen types with minimal sequence bias. LC-MS/MS analysis of enriched mouse urine revealed an average of 383 collagenous peptide fragments per sample (compared to 34 for unenriched sample) which could be mapped to all types of mouse collagens in the SwissProt database including FACITs and MACITs. Hierarchical clustering of a selected panel of the detected fragments separated osteopenic mice from healthy mice. The results demonstrate dimeric CHP’s ability to enrich collagen fragments from biological fluid and its potential to aid peptidomics-based disease detection and biomarker discovery. [Raw mass spectrometry files have been deposited to the MassIVE repository (massive.ucsd.edu, MSV000085372, doi 10.25345/C5N12H)].
- Published
- 2020
73. YIA20-003: CD229 CAR T Cells Eliminate Multiple Myeloma and Tumor Propagating Cells but Show Limited Targeting of Normal T Cells
- Author
-
Sandra D. Scherer, Alana L. Welm, Sara Yousef, Tim Luetkens, Rodney R. Miles, Yasmina Noubia Abdiche, Sabarinath Venniyil Radhakrishnan, K. David Li, Djordje Atanackovic, Erica R. Vander Mause, William Matsui, Patricia Davis, Jens Panse, and Michael L. Olson
- Subjects
Oncology ,business.industry ,Cancer research ,Medicine ,Car t cells ,business ,medicine.disease ,Multiple myeloma - Published
- 2020
74. Low-affinity CAR T cells exhibit reduced trogocytosis, preventing fratricide and antigen-negative tumor escape while preserving anti-tumor activity
- Author
-
Michael L. Olson, Erica R. Vander Mause, Sabarinath V. Radhakrishnan, Joshua D. Brody, Aaron P. Rapoport, Alana L. Welm, Djordje Atanackovic, and Tim Luetkens
- Abstract
Chimeric antigen receptor (CAR) T cells using the high-affinity CD19 binding domain FMC63 are an effective treatment for patients with relapsed and aggressive B cell lymphoma. However, antigen loss and poor CAR T cell persistence remain common causes for relapse in these patients. Using primary patient samples, we now show that FMC63-based CAR T cells confer rapid antigen loss in all major tumor types currently approved for treatment with CD19 CAR T cells via trogocytosis, the stripping of antigen from tumor cells by CAR T cells. We show that CAR T cell-mediated trogocytosis can be dramatically reduced across a wide range of B cell malignancies by replacing FMC63 with a low affinity CD19 antibody. This reduction in trogocytosis does not alter the direct anti-tumor activity of CD19 CAR T cells but prevents the emergence of antigen-negative tumor cells and significantly increases CAR T cell viability by reducing fratricide of CD19 CAR T cells following trogocytosis.TEASERA reduction in CAR affinity does not affect tumor killing but prolongs T cell persistence and prevents antigen-negative tumor escape.
- Published
- 2021
75. Abstract A55: Defining the immune milieu in short-form RON-mediated tumor clearance in breast cancer bone metastasis
- Author
-
Clint H Valencia, Jaime Fornetti, and Alana L Welm
- Subjects
Cancer Research ,Immunology - Abstract
The bone is the most common site of metastasis and the usual first site of recurrence in metastatic breast cancer. Bone metastasis is incurable and causes severe pain with significant bone loss in breast cancer patients. Current treatments for bone metastasis aim to diminish bone loss; however, most patients still progress on current therapies, making it critical to identify new therapeutic targets. We have identified the receptor Recepteur d'Origine Nantais (RON) as a target for treating metastatic breast cancer. RON is the receptor for the macrophage stimulating protein (MSP) and has both tumor-intrinsic and -extrinsic roles in metastasis. The gene encoding RON (MST1R in human or Stk in mouse) gives rise to two transcripts coding for full-length RON (FL-RON) and short-form RON (SF-RON) isoforms, where SF-RON lacks the ligand-binding domain for MSP and is constitutively active. In mouse breast cancer lung metastasis models, deletion of kinase activity from both isoforms of RON in the host nearly eradicates tumors through increased anti-tumor immunity. More recently, we identified SF-RON as the main isoform mediating tumor-associated immunity in these models, with loss of SF-RON promoting increased anti-tumor T cell activity. In the bone, loss of kinase activity from both isoforms of RON protects against bone loss, but its role in anti-tumor immunity is unknown. We now aim to delineate the role of SF-RON-mediated anti-tumor immune responses in the bone, where immunotherapy has been less effective. In SF-RON knockout mice (RonSF-/-), tumor cells injected into the bone grow for a short time but are undetectable by endpoint. Preliminary analyses suggest the importance of immune-mediated clearance in the loss of tumors in RonSF-/- mice as depletion of CD8+ T cells rescued tumor growth by approximately 50%. To gain insight into the role of immune cell populations in this anti-tumor response, we utilized immunohistochemistry and flow cytometry to characterize specific immune cells in bone metastasis. Initial results suggest increased infiltration of T cells, B cells, and NK cells into the tumors in the bones of RonSF-/- mice compared to wild-type controls. Notably, these immune cell populations are all known to play a significant role in the anti-tumor response in other models. Future work will utilize depletion studies to further investigate the role of T cells, B Cells, and NK cells in SF-RON-mediated tumor clearance in the bone. This work will help elucidate a role for host SF-RON in breast cancer bone metastasis and its potential to be a potent immunotherapy target for the treatment of bone metastasis. Citation Format: Clint H Valencia, Jaime Fornetti, Alana L Welm. Defining the immune milieu in short-form RON-mediated tumor clearance in breast cancer bone metastasis [abstract]. In: Proceedings of the AACR Special Conference: Tumor Immunology and Immunotherapy; 2022 Oct 21-24; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2022;10(12 Suppl):Abstract nr A55.
- Published
- 2022
76. 870 Targeting GCN2 kinase-driven stress response inactivation to restore tumor immunity in metastatic triple negative breast cancer
- Author
-
Zhaoliang Li, Kyle Medley, David J. Bearss, Hariprasad Vankayalapati, Alana L. Welm, and Dongqing Yan
- Subjects
Pharmacology ,Cancer Research ,business.industry ,Kinase ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Tumor immunity ,Fight-or-flight response ,Oncology ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Medicine ,business ,Triple-negative breast cancer ,RC254-282 - Abstract
BackgroundPatients with PD-L1-positive metastatic triple-negative breast cancers (mTNBC) who have been treated with atezolizumab+nab-paclitaxel had a clinically meaningful overall survival extension of 9.5 months compared to nab-paclitaxel alone, although overall survival in overall population was not statistically significant. Unlike many other cancers, immunotherapy for breast cancer has had limited success, due to the fact that there are very few T cells in the tumor microenvironment of mTNBC patients. Identifying ways to boost immunotherapy responses could change the paradigm of mTNBC, a disease still difficult to treat. The highly proliferative nature of tumor cells, along with infiltration of myeloid cells into the tumors, leads to depletion of nutrients such as functional/natural amino acids. This metabolically stressful milieu causes activation of nutrient stress pathways, autophagy, and repressed immune responses. A key meditator of this nutrient stress pathway is a cytoplasmic Ser/Thr protein kinase called General Control Nonderepressible 2 (GCN2), also called EIF2AK4. GCN2 switches on following reduction of amino acids, and its activity results in T cell inactivation, T cell death, regulatory T cell expansion, and the potentiation of myeloid-derived suppressor cells (MDSCs).MethodsWe have developed and synthesized a series of novel small molecule immunotherapeutic agents that reversibly bind to GCN2 kinase, competitively block the ATP site, and elicit pharmacological responses in immune cells and in breast cancer cells.ResultsGCN2 cell-free kinase binding, kinome selectivity, pGCN2, pEIF2α, ATF-4 phosphorylation inhibition assays were performed. We confirmed on-target efficacy and tested the potency of our lead GCN2 inhibitor HCI-1046. HCI-1046 demonstrated potent activity, with an IC50 of 36 nM in inhibiting GCN2 kinase and exhibited cellular efficacy with an IC50 of 0.1 to 1.0 μM range. Our preliminary results support the hypothesis that the inhibition of GCN2 reinstates anti-tumor immunity and blocks tumor progression in breast cancer models. In vivo PK studies of HCI-1046 in rodents showed excellent PK properties; 55% oral bioavailability, low clearance, and >5 hour half-life.ConclusionsThus, HCI-1046 is nominated as a pre-clinical agent. Additional data regarding evaluation of the effects of HCI-1046 on the MDSC-suppressive function on T cells using ELISpot assays with breast cancer patient samples, and mouse model efficacy studies will be discussed.ReferencesEkiz HA, Lai SA, Gundlapalli H, Haroun F, Williams MA, Welm AL. Inhibition of RON kinase potentiates anti-CTLA-4 immunotherapy to shrink breast tumors and prevent metastatic outgrowth. Oncoimmunology 2018;7(9):e1480286.Toogood PL. Small molecule immuno-oncology therapeutic agents. Bioorg Med Chem Lett 2018;28(3):319–329.Ravindran R, Loebbermann J, Nakaya HI, Khan N, Ma H, Gama L, Machiah DK, Lawson B, Hakimpour P, Wang YC, Li S, Sharma P, Kaufman RJ, Martinez J, Pulendran B. The amino acid sensor GCN2 controls gut inflammation by inhibiting inflammasome activation. Nature 2016;531(7595):523–527.Brazeau JF, Rosse G. Triazolo[4,5-d]pyrimidine derivatives as inhibitors of GCN2. ACS Med Chem Lett 2014;5(4):282–3.
- Published
- 2021
77. PDXNet Portal: Patient-Derived Xenograft model, data, workflow, and tool discovery
- Author
-
Li Ding, Michael Davies, Xing Yi Woo, Jeffrey H. Chuang, Carol J. Bult, Chong-Xian Pan, Brian S. White, Yvonne A. Evrard, Alana L. Welm, Christian Frech, Manisha Ray, Ramaswamy Govindan, Salma Kaochar, John DiGiovanna, Paul Lott, Anuj Srivastava, Luis G. Carvajal-Carmona, Bryan E. Welm, Steven B. Neuhauser, Tiffany A. Wallace, Sai Lakshmi Subramanian, Meenherd Herlyn, Brandi N. Davis-Dusenbery, Phillip Webster, Dennis A. Dean, James H. Doroshow, Nicholas Mitsiades, Soner Koc, Jeffrey W. Grover, Jack A. Roth, Michael T. Lewis, Sara Seepo, Funda Meric-Bernstam, Shunquang Li, Michael Lloyd, Moon S. Chen, Peter N. Robinson, Jeffrey A. Moscow, and Brian J. Sanderson
- Subjects
Public access ,World Wide Web ,Data processing ,Service (systems architecture) ,Workflow ,Computer science ,business.industry ,Genomics ,Cloud computing ,business ,Data resources ,Tumor xenograft - Abstract
We created the PDX Network (PDXNet) Portal (https://portal.pdxnetwork.org/) to centralize access to the National Cancer Institute-funded PDXNet consortium resources (i.e., PDX models, sequencing data, treatment response data, and bioinformatics workflows), to facilitate collaboration among researchers, and to make resources easily available for research. The portal includes sections for resources, analysis results, metrics for PDXNet activities, data processing protocols, and training materials for processing PDX data.The initial portal release highlights PDXNet model and data resources, including 334 new models across 33 cancer types. Tissue samples of these models were deposited in the NCI’s Patient-Derived Model Repository (PDMR) for public access. These models have 2,822 associated sequencing files from 873 samples across 307 patients, which are hosted on the Cancer Genomics Cloud powered by Seven Bridges and the NCI Cancer Data Service for long-term storage and access with dbGaP permissions. The portal also includes results from standardized analysis workflows on PDXNet sequencing files and PDMR data (2,594 samples from 463 patients across 78 disease types). These 15 analysis workflows for whole-exome and RNA-Seq data are freely available, robust, validated, and standardized.The model and data lists will grow substantially over the next two years and will be continuously updated as new data are available. PDXNet models support multi-agent treatment studies, determination of sensitivity and resistance mechanisms, and preclinical trials. The PDXNet portal is a centralized location for these data and resources, which we expect to be of significant utility for the cancer research community.
- Published
- 2021
78. An immune-humanized patient-derived xenograft model of estrogen-independent, hormone receptor positive metastatic breast cancer
- Author
-
Maihi Fujita, Alana L. Welm, Zheqi Li, Steffi Oesterreich, Yoko S. DeRose, Jennifer Toner, Alessandra I. Riggio, Ahmed A. Samatar, Fadi Haroun, Ling Zhao, H. Atakan Ekiz, and Sandra D. Scherer
- Subjects
ER+ PDX ,Antigens, CD34 ,Breast Neoplasms ,Mice, Transgenic ,Mice, SCID ,Estrogen supplementation ,Mice ,Breast cancer ,Surgical oncology ,Mice, Inbred NOD ,medicine ,Tumor Microenvironment ,Animals ,Humans ,Humanized PDX ,skin and connective tissue diseases ,RC254-282 ,Tumor microenvironment ,business.industry ,Y537S ,Estrogen Receptor alpha ,Hematopoietic Stem Cell Transplantation ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cancer ,ER+ metastatic breast cancer ,Immune-humanization ,Estrogens ,medicine.disease ,Hematopoietic Stem Cells ,Metastatic breast cancer ,ESR1 mutation ,Xenograft Model Antitumor Assays ,Breast cancer tumor microenvironment ,Disease Models, Animal ,medicine.anatomical_structure ,Receptors, Estrogen ,Tumor progression ,Drug Resistance, Neoplasm ,Mutation ,Cancer research ,bacteria ,Heterografts ,Female ,Bone marrow ,business ,Estrogen receptor alpha ,Research Article ,Endocrine resistance model - Abstract
Background Metastatic breast cancer (MBC) is incurable, with a 5-year survival rate of 28%. In the USA, more than 42,000 patients die from MBC every year. The most common type of breast cancer is estrogen receptor-positive (ER+), and more patients die from ER+ breast cancer than from any other subtype. ER+ tumors can be successfully treated with hormone therapy, but many tumors acquire endocrine resistance, at which point treatment options are limited. There is an urgent need for model systems that better represent human ER+ MBC in vivo, where tumors can metastasize. Patient-derived xenografts (PDX) made from MBC spontaneously metastasize, but the immunodeficient host is a caveat, given the known role of the immune system in tumor progression and response to therapy. Thus, we attempted to develop an immune-humanized PDX model of ER+ MBC. Methods NSG-SGM3 mice were immune-humanized with CD34+ hematopoietic stem cells, followed by engraftment of human ER+ endocrine resistant MBC tumor fragments. Strategies for exogenous estrogen supplementation were compared, and immune-humanization in blood, bone marrow, spleen, and tumors was assessed by flow cytometry and tissue immunostaining. Characterization of the new model includes assessment of the human tumor microenvironment performed by immunostaining. Results We describe the development of an immune-humanized PDX model of estrogen-independent endocrine resistant ER+ MBC. Importantly, our model harbors a naturally occurring ESR1 mutation, and immune-humanization recapitulates the lymphocyte-excluded and myeloid-rich tumor microenvironment of human ER+ breast tumors. Conclusion This model sets the stage for development of other clinically relevant models of human breast cancer and should allow future studies on mechanisms of endocrine resistance and tumor-immune interactions in an immune-humanized in vivo setting.
- Published
- 2021
79. Heterogeneity in Metastatic Potential of Cancer Cells Is Revealed En Masse
- Author
-
Alana L. Welm and Pavitra Viswanath
- Subjects
0301 basic medicine ,Cancer Research ,business.industry ,Lipid metabolism ,Biology ,medicine.disease ,Metastasis ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Text mining ,Oncology ,Breast cancer cell line ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,medicine ,business ,Tropism - Abstract
A new study in Nature determines metastatic tropism in xenograft mouse models. This results in a metastasis map for 21 tumor types, the utility of which is demonstrated by identifying lipid metabolism to be uniquely altered in breast cancer cell lines that metastasize to the brain.
- Published
- 2021
80. Laterally transferred macrophage mitochondria act as a signaling source promoting cancer cell proliferation
- Author
-
Chelsea U Kidwell, Gregory S. Olson, Jarrod S. Johnson, Alana L. Welm, Soorya Pradeep, Sandra D. Scherer, Joseph R. Casalini, Minna Roh-Johnson, Jared Rutter, Daniel Greiner, and Thomas A. Zangle
- Subjects
Membrane potential ,chemistry.chemical_classification ,Reactive oxygen species ,Chemistry ,Cancer cell proliferation ,Cancer cell ,Erk signaling ,Ros signaling ,Macrophage ,Mitochondrion ,Cell biology - Abstract
Lateral transfer of mitochondria occurs in many physiological and pathological conditions. Given that mitochondria provide essential energy for cellular activities, mitochondrial transfer is currently thought to promote the rescue of damaged cells. We report that mitochondrial transfer occurs between macrophages and breast cancer cells, leading to increased cancer cell proliferation. Unexpectedly, transferred macrophage mitochondria are dysfunctional, lacking mitochondrial membrane potential. Rather than performing essential mitochondrial activities, transferred mitochondria accumulate reactive oxygen species which activates ERK signaling, indicating that transferred mitochondria act as a signaling source that promotes cancer cell proliferation. We also demonstrate that pro-tumorigenic M2-like macrophages exhibit increased mitochondrial transfer to cancer cells. Collectively, our findings reveal how mitochondrial transfer is regulated and leads to sustained functional changes in recipient cells.One-Sentence SummaryLateral transfer of macrophage mitochondria acts as a ROS signaling source, regulating cancer cell proliferation through ERK signaling.
- Published
- 2021
81. Functional precision oncology: Testing tumors with drugs to identify vulnerabilities and novel combinations
- Author
-
Anthony Letai, Patrick Bhola, and Alana L. Welm
- Subjects
Cancer Research ,Oncology ,Pharmaceutical Preparations ,Neoplasms ,Mutation ,Biomarkers, Tumor ,Humans ,Precision Medicine ,Medical Oncology ,Article - Abstract
Functional precision medicine is a strategy whereby live tumor cells from affected individuals are directly perturbed with drugs to provide immediately translatable, personalized information to guide therapy. The heterogeneity of human cancer has led to the realization that personalized approaches are needed to improve treatment outcomes. Precision oncology has traditionally used static features of the tumor to dictate which therapies should be used. Static features can include expression of key targets or genomic analysis of mutations to identify therapeutically targetable "drivers." Although a surprisingly small proportion of individuals derive clinical benefit from the static approach, functional precision medicine can provide additional information regarding tumor vulnerabilities. We discuss emerging technologies for functional precision medicine as well as limitations and challenges in using these assays in the clinical trials that will be necessary to determine whether functional precision medicine can improve outcomes and eventually become a standard tool in clinical oncology.
- Published
- 2021
82. Evaluation of FGFR targeting in breast cancer through interrogation of patient-derived models
- Author
-
Elizabeth V. Nguyen, Diana J. Micati, Genevieve Kerr, Mun N. Hui, Elgene Lim, Nicole J Chew, Tali Lang, Peter Gregory, Catriona McLean, Niantao Deng, Gary Richardson, Terry C C Lim Kam Sian, Helen E. Abud, Thierry Jarde, Sung-Young Shin, Melissa Vereker, Alexander Swarbrick, Jessica Yang, Roger J. Daly, Alana L. Welm, Rachel S. Lee, and Tim Nottle
- Subjects
medicine.medical_treatment ,Breast Neoplasms ,Triple Negative Breast Neoplasms ,Biology ,Models, Biological ,Targeted therapy ,Mice ,Breast cancer ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,Phosphorylation ,Precision Medicine ,Protein Kinase Inhibitors ,Oncogene ,RC254-282 ,Kinase ,Fibroblast growth factor receptor 1 ,Phosphoproteomics ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cancer ,Precision oncology ,medicine.disease ,Receptors, Fibroblast Growth Factor ,Xenograft Model Antitumor Assays ,Fibroblast growth factor receptor ,DNA-Binding Proteins ,Organoids ,Mutation ,Cancer research ,SKI proto-oncogene ,Tyrosine kinase ,Research Article - Abstract
Background Particular breast cancer subtypes pose a clinical challenge due to limited targeted therapeutic options and/or poor responses to the existing targeted therapies. While cell lines provide useful pre-clinical models, patient-derived xenografts (PDX) and organoids (PDO) provide significant advantages, including maintenance of genetic and phenotypic heterogeneity, 3D architecture and for PDX, tumor–stroma interactions. In this study, we applied an integrated multi-omic approach across panels of breast cancer PDXs and PDOs in order to identify candidate therapeutic targets, with a major focus on specific FGFRs. Methods MS-based phosphoproteomics, RNAseq, WES and Western blotting were used to characterize aberrantly activated protein kinases and effects of specific FGFR inhibitors. PDX and PDO were treated with the selective tyrosine kinase inhibitors AZD4547 (FGFR1-3) and BLU9931 (FGFR4). FGFR4 expression in cancer tissue samples and PDOs was assessed by immunohistochemistry. METABRIC and TCGA datasets were interrogated to identify specific FGFR alterations and their association with breast cancer subtype and patient survival. Results Phosphoproteomic profiling across 18 triple-negative breast cancers (TNBC) and 1 luminal B PDX revealed considerable heterogeneity in kinase activation, but 1/3 of PDX exhibited enhanced phosphorylation of FGFR1, FGFR2 or FGFR4. One TNBC PDX with high FGFR2 activation was exquisitely sensitive to AZD4547. Integrated ‘omic analysis revealed a novel FGFR2-SKI fusion that comprised the majority of FGFR2 joined to the C-terminal region of SKI containing the coiled-coil domains. High FGFR4 phosphorylation characterized a luminal B PDX model and treatment with BLU9931 significantly decreased tumor growth. Phosphoproteomic and transcriptomic analyses confirmed on-target action of the two anti-FGFR drugs and also revealed novel effects on the spliceosome, metabolism and extracellular matrix (AZD4547) and RIG-I-like and NOD-like receptor signaling (BLU9931). Interrogation of public datasets revealed FGFR2 amplification, fusion or mutation in TNBC and other breast cancer subtypes, while FGFR4 overexpression and amplification occurred in all breast cancer subtypes and were associated with poor prognosis. Characterization of a PDO panel identified a luminal A PDO with high FGFR4 expression that was sensitive to BLU9931 treatment, further highlighting FGFR4 as a potential therapeutic target. Conclusions This work highlights how patient-derived models of human breast cancer provide powerful platforms for therapeutic target identification and analysis of drug action, and also the potential of specific FGFRs, including FGFR4, as targets for precision treatment.
- Published
- 2021
83. Protective autophagy elicited by RAF→MEK→ERK inhibition suggests a treatment strategy for RAS-driven cancers
- Author
-
Mona Foth, Courtney C. Cavalieri, Lance D. Burrell, Stephanie L. Cutler, Conan G. Kinsey, Kajsa E. Affolter, Jill E. Shea, Amelie M. Boespflug, Kaitrin M. Rehbein, Alana L. Welm, Jeffrey T. Yap, Bryan E. Welm, Michael T. Seipp, Sophia S. Schuman, Jonathan Whisenant, G. Weldon Gilcrease, Amanda Truong, David H. Lum, Katrin P. Guillen, Courtney L. Scaife, Martin McMahon, Eric L. Snyder, and Soledad A. Camolotto
- Subjects
Proto-Oncogene Proteins B-raf ,0301 basic medicine ,MAPK/ERK pathway ,CA-19-9 Antigen ,MAP Kinase Signaling System ,Pyridones ,Mice, SCID ,Pyrimidinones ,medicine.disease_cause ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Pancreatic cancer ,Autophagy ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,Mitogen-Activated Protein Kinase Kinases ,Trametinib ,business.industry ,Melanoma ,Cancer ,Chloroquine ,General Medicine ,medicine.disease ,Xenograft Model Antitumor Assays ,3. Good health ,Pancreatic Neoplasms ,Editorial Commentary ,030104 developmental biology ,030220 oncology & carcinogenesis ,ras Proteins ,Cancer research ,KRAS ,Signal transduction ,business - Abstract
Pancreatic ductal adenocarcinoma (PDA) was responsible for ~ 44,000 deaths in the United States in 2018 and is the epitome of a recalcitrant cancer driven by a pharmacologically intractable oncoprotein, KRAS1–4. Downstream of KRAS, the RAF→MEK→ERK signaling pathway plays a central role in pancreatic carcinogenesis5. However, paradoxically, inhibition of this pathway has provided no clinical benefit to patients with PDA6. Here we show that inhibition of KRAS→RAF→MEK→ERK signaling elicits autophagy, a process of cellular recycling that protects PDA cells from the cytotoxic effects of KRAS pathway inhibition. Mechanistically, inhibition of MEK1/2 leads to activation of the LKB1→AMPK→ULK1 signaling axis, a key regulator of autophagy. Furthermore, combined inhibition of MEK1/2 plus autophagy displays synergistic anti-proliferative effects against PDA cell lines in vitro and promotes regression of xenografted patient-derived PDA tumors in mice. The observed effect of combination trametinib plus chloroquine was not restricted to PDA as other tumors, including patient-derived xenografts (PDX) of NRAS-mutated melanoma and BRAF-mutated colorectal cancer displayed similar responses. Finally, treatment of a patient with PDA with the combination of trametinib plus hydroxychloroquine resulted in a partial, but nonetheless striking disease response. These data suggest that this combination therapy may represent a novel strategy to target RAS-driven cancers. Targeted inhibition of RAF–MEK–ERK signaling induces autophagy through the LKB1–AMPK axis, creating a therapeutic vulnerability that can be exploited for treating patients with pancreatic cancer and potentially other RAS-mutant tumors.
- Published
- 2019
84. Abstract 1014: A framework for research scientists to include patient advocates in cancer research
- Author
-
Hillary Stires, Igor Bado, Thelma Brown, Martha Carlson, Isaac S. Chan, Gloria V. Echeverria, Andrew J. Ewald, Carla Lloyd, Julia Maues, Steffi Oesterreich, Robert N. Riter, Kelly Shanahan, Alana L. Welm, and Josh Newby
- Subjects
Cancer Research ,Oncology - Abstract
The inclusion of patient advocates in basic cancer research has emerged as a valuable practice to ensure research is intentional, to support effective communication with broader audiences, and to directly connect researchers with those whom they are striving to help. Despite this value, many researchers do not work with patient advocates. To understand why and build a roadmap for more engagement in the future, we hosted a workshop with patient advocates and research scientists then presented findings and discussed further at an international conference. We acknowledged four main barriers: 1) It is not clear to everyone why patient advocates should be included in research, 2) Researchers are worried about saying the wrong thing, 3) Researchers do not know where to meet patient advocates, and 4) Researchers do not know how to include patient advocates in research. We identified best practices from various organizations and opportunities to overcome these barriers in the short- and long-term. This is the first time a multi-stakeholder group has come together to provide ways to support research scientists with overcoming barriers to regularly working with patient advocates. Ultimately, these relationships will improve cancer research and more quickly accomplish our collective goal of improving lives of those who have been diagnosed with cancer. Citation Format: Hillary Stires, Igor Bado, Thelma Brown, Martha Carlson, Isaac S. Chan, Gloria V. Echeverria, Andrew J. Ewald, Carla Lloyd, Julia Maues, Steffi Oesterreich, Robert N. Riter, Kelly Shanahan, Alana L. Welm, Josh Newby. A framework for research scientists to include patient advocates in cancer research [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 1014.
- Published
- 2022
85. Abstract 2723: Model-based cancer therapy selection by linking tumor vulnerabilities to drug mechanism
- Author
-
Szabolcs Tarapcsak, Yi Qiao, Xiaomeng Huang, Tony Di Sera, Matthew H. Bailey, Bryan E. Welm, Alana L. Welm, and Gabor T. Marth
- Subjects
Cancer Research ,Oncology - Abstract
Selection of the treatments most likely to combat a patient’s tumor is a central aim of precision oncology. We are currently developing a functional precision oncology program at the University of Utah and Huntsman Cancer Institute that combines the multi-omic characterization of a patient's tumor with the functional screening of relevant drug candidates in patient-derived organoid tumor models to identify which drugs are uniquely capable of combating the patient’s cancer. Here we present our model-based approach for utilizing the multi-omic tumor data to computationally predict the patient’s response to each drug. In this approach, we first identify the genomic and transcriptomic vulnerabilities of the tumor as genes harboring somatic DNA mutations or copy number changes (from paired tumor/normal WGS/WES DNA sequencing data), as well as genes whose expression levels have been significantly altered in the tumor (based on bulk or single-cell RNA sequencing data). Second, using targeting information from drug-gene interaction databases (DGIdb) we compile a list of genes targeted by each drug relevant in the patient’s treatment. Third, we utilize gene interaction database knowledge (KEGG) to construct a gene interaction graph to link each drug’s gene targets with all of the tumor’s vulnerability genes. We then identify all gene interaction paths connecting specific drug target genes with specific vulnerability genes; and score each path and combine all path-specific scores for the target-vulnerability gene pair. Subsequently, we further combine all pairwise scores across all drug target genes and all tumor vulnerability genes and determine the statistical significance of the resulting score by sampling the background distribution of such scores across random drugs, target genes, and vulnerability genes. We have applied our algorithm to predict drug response in advanced breast cancer patients as well as using publicly available tumor cell line data (GDSC2). We have found a high level of concordance between our computational prediction and organoid/cell line screening results, clearly separating sensitive and non-sensitive models. Using Bayesian probability, we compare the drug-specific score distributions of sensitive and non-sensitive models and are able to identify sensitive cell lines/patients with high accuracy. By encapsulating available information on direct gene-gene interactions, the drug’s direct gene targets and the collected omic vulnerabilities of the tumor, our model is not only capable of predicting sensitivity to both targeted and chemotherapy agents, but can also provide a mechanistic understanding for the targeting of the tumor. Our approach will be validated and fine-tuned on a large cohort of breast and brain cancer patients as well as patient PDX models, interrogating gene-target interactions to identify novel relevant target genes/pathways. Citation Format: Szabolcs Tarapcsak, Yi Qiao, Xiaomeng Huang, Tony Di Sera, Matthew H. Bailey, Bryan E. Welm, Alana L. Welm, Gabor T. Marth. Model-based cancer therapy selection by linking tumor vulnerabilities to drug mechanism [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 2723.
- Published
- 2022
86. A breast cancer patient-derived xenograft and organoid platform for drug discovery and precision oncology
- Author
-
Ward Jh, Randy L. Jensen, Ling Zhao, Greenland Ja, Maihi Fujita, Rosenthal R, Saundra S. Buys, Anna C. Beck, Guoying Wang, Wadsworth Me, Emilio Cortes-Sanchez, Sandra D. Scherer, Chieh-Hsiang Yang, Katrin P. Guillen, Jeffrey H. Chuang, Cindy B. Matsen, Zheqi Li, Kristopher Berrett, Jeffery M. Vahrenkamp, David H. Lum, Alana L. Welm, Michael T. Lewis, Toner J, Jason Gertz, Chu Z, Poretta Jm, Yoko S. DeRose, Lacey E. Dobrolecki, Gabor T. Marth, Katherine E. Varley, Rachel E. Factor, Andrew Butterfield, Bryan E. Welm, Xiaomeng Huang, Edward W. Nelson, Matthew H. Bailey, Pathi Ss, Xing Yi Woo, Yi Qiao, Christos Vaklavas, Steffi Oesterreich, and Kevin B. Jones
- Subjects
Drug ,Oncology ,medicine.medical_specialty ,Drug discovery ,business.industry ,media_common.quotation_subject ,Cancer ,Precision medicine ,medicine.disease ,Breast cancer ,Drug development ,In vivo ,Internal medicine ,Medicine ,business ,Triple-negative breast cancer ,media_common - Abstract
Model systems that recapitulate the complexity of human tumors and the reality of variable treatment responses are urgently needed to better understand cancer biology and to develop more effective cancer therapies. Here we report development and characterization of a large bank of patient-derived xenografts (PDX) and matched organoid cultures from tumors that represent some of the greatest unmet needs in breast cancer research and treatment. These include endocrine-resistant, treatment-refractory, and metastatic breast cancers and, in some cases, multiple tumor collections from the same patients. The models can be grown long-term with high fidelity to the original tumors. We show that development of matched PDX and PDX-derived organoid (PDxO) models facilitates high-throughput drug screening that is feasible and cost-effective, while also allowing in vivo validation of results. Our data reveal consistency between drug screening results in organoids and drug responses in breast cancer PDX. Moreover, we demonstrate the feasibility of using these patient-derived models for precision oncology in real time with patient care, using a case of a triple negative breast cancer with early metastatic recurrence as an example. Our results uncovered an FDA-approved drug with high efficacy against the models. Treatment with the PDxO-directed therapy resulted in a complete response for the patient and a progression-free survival period more than three times longer than her previous therapies. This work provides valuable new methods and resources for functional precision medicine and drug development for human breast cancer.Graphical Abstract
- Published
- 2021
87. An embryonic diapause-like adaptation with suppressed Myc activity enables tumor treatment persistence
- Author
-
Dhvanir Kansara, Eugen Dhimolea, Pallavi Awate, Alana L. Welm, Jordan Bryan, Brian J. Glassner, Zhiyi Liu, Shruti Sharma, Juliette Bouyssou, Rinath Jeselsohn, Aziz Al'Khafaji, Huihui Tang, Myles Brown, Nathanael S. Gray, Joseline Raja, Boris Bartholdy, Cihangir Duy, Aedín C. Culhane, Yu Chen, Xiang Weng, Ricardo De Matos Simoes, Jennifer Roth, Irene Georgakoudi, Dong Gao, Constantine S. Mitsiades, Ari Melnick, Ryosuke Shirasaki, Aviad Tsherniak, Francisca Vazquez, Michal Scheffer, and Samantha Bender
- Subjects
0301 basic medicine ,Cancer Research ,BRD4 ,Transcription, Genetic ,Antineoplastic Agents ,Apoptosis ,Biology ,Article ,Cell Line ,Proto-Oncogene Proteins c-myc ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Kinase ,Cancer ,Embryo, Mammalian ,medicine.disease ,Adaptation, Physiological ,Cyclin-Dependent Kinase 9 ,Diapause ,Up-Regulation ,HEK293 Cells ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,MCF-7 Cells ,Cancer research ,Female ,Cyclin-dependent kinase 9 ,Embryonic diapause ,Transcription Factors - Abstract
Treatment-persistent residual tumors impede curative cancer therapy. To understand this cancer cell state we generate models of treatment persistence that simulate the residual tumors. We observe that treatment-persistent tumor cells in organoids, xenografts and in cancer patients adopt a distinct and reversible transcriptional program resembling that of embryonic diapause, a dormant stage of suspended development triggered by stress and associated with suppressed Myc activity and overall biosynthesis. In cancer cells, depleting Myc or inhibiting Brd4, a Myc transcriptional co-activator, abrogates drug cytotoxicity through a dormant diapause-like adaptation with reduced apoptotic priming. Conversely, inducible Myc upregulation enhances acute chemotherapeutic activity. Maintaining residual cells in dormancy after chemotherapy by inhibiting Myc activity, or interfering with the diapause-like adaptation by inhibiting cyclin-dependent kinase 9 (CDK9), represent potential therapeutic strategies against chemotherapy-persistent tumor cells. Our study demonstrates that cancer co-opts a mechanism similar to diapause with adaptive inactivation of Myc to persist during treatment.
- Published
- 2021
88. CDK/cyclin dependencies define extreme cancer cell-cycle heterogeneity and collateral vulnerabilities
- Author
-
Erik S. Knudsen, Vishnu Kumarasamy, Ram Nambiar, Joel D. Pearson, Paris Vail, Hanna Rosenheck, Jianxin Wang, Kevin Eng, Rod Bremner, Daniel Schramek, Seth M. Rubin, Alana L. Welm, and Agnieszka K. Witkiewicz
- Subjects
Neoplasms ,Cell Cycle ,Cyclin-Dependent Kinase 4 ,Humans ,Cell Cycle Proteins ,Cell Division ,Cyclin-Dependent Kinase Inhibitor p27 ,Cyclin-Dependent Kinases ,General Biochemistry, Genetics and Molecular Biology - Abstract
Progression through G1/S phase of the cell cycle is coordinated by cyclin-dependent kinase (CDK) activities. Here, we find that the requirement for different CDK activities and cyclins in driving cancer cell cycles is highly heterogeneous. The differential gene requirements associate with tumor origin and genetic alterations. We define multiple mechanisms for G1/S progression in RB-proficient models, which are CDK4/6 independent and elicit resistance to FDA-approved inhibitors. Conversely, RB-deficient models are intrinsically CDK4/6 independent, but exhibit differential requirements for cyclin E. These dependencies for CDK and cyclins associate with gene expression programs that denote intrinsically different cell-cycle states. Mining therapeutic sensitivities shows that there are reciprocal vulnerabilities associated with RB1 or CCND1 expression versus CCNE1 or CDKN2A. Together, these findings illustrate the complex nature of cancer cell cycles and the relevance for precision therapeutic intervention.
- Published
- 2022
89. ScRNA-seq reveals tumor microenvironment remodeling induced by local intervention-based immunotherapy
- Author
-
Xiao Hong Sun, Jason R. Krawic, Delaney Medcalf, Christa I. DeVette, Kaili Liu, Alana L. Welm, Wei R. Chen, William H. Hildebrand, Ashley R. Hoover, and Connor L. West
- Subjects
Tumor microenvironment ,Immune system ,Myeloid ,medicine.anatomical_structure ,Cytokine ,medicine.medical_treatment ,T cell ,Cancer research ,medicine ,Tumor necrosis factor alpha ,Immunotherapy ,Biology ,Proinflammatory cytokine - Abstract
Laser immunotherapy (LIT) combines local photothermal therapy (PTT), to disrupt tumor homeostasis and release tumor antigens, and an intratumorally administered immunostimulant, N-dihydrogalactochitosan (GC), to induce antitumor immune responses. We performed single-cell RNA sequencing on tumor-infiltrating leukocytes of MMTV-PyMT mouse mammary tumors to characterize LIT-induced myeloid and lymphoid compartment remodeling. Analysis of 49,380 single cell transcriptomes from different treatment groups revealed that proinflammatory IFNα, IFNγ, and TNFα cytokine signaling pathways were enriched in both lymphoid and myeloid cells isolated from LIT-treated tumors. The CD4+ and CD8+ T cells in LIT treated tumors resided in an activated state while immune cells in untreated and PTT-treated tumors remained in a neutral/resting state. Additionally, monocytes recruited into the LIT-treated tumors were driven towards proinflammatory M1-like macrophage phenotypes or monocyte-derived dendritic cells. Our results reveal that LIT prompts immunological remodeling of the tumor microenvironment by initiating broad proinflammatory responses to drive antitumor immunity.STATEMENT OF SIGNIFICANCETranscriptome profiling of tumor infiltrating leukocytes revealed that localized laser immunotherapy (LIT) greatly enhanced antitumor T cell activity by promoting proinflammatory myeloid cell responses within the tumor microenvironment. This manuscript demonstrates that LIT broadly stimulates antitumor immunity and has great potential to synergize with current immunotherapies to increase their efficacy.
- Published
- 2020
90. Tumoural activation of TLR3-SLIT2 axis in endothelium drives metastasis
- Author
-
Alana L. Welm, Bernardo Tavora, Olav Olsen, Mahan Sadjadi, Ji-Young Kim, Kai J. Wessel, Xuhang Liu, Marc Missmahl, Sohail F. Tavazoie, Simon Ruffing, Tobias Mederer, Hani Goodarzi, and Benjamin N. Ostendorf
- Subjects
0301 basic medicine ,Male ,Lung Neoplasms ,Metastasis ,Mice ,Double-Stranded ,0302 clinical medicine ,Immunologic ,Receptors ,Tumor Cells, Cultured ,2.1 Biological and endogenous factors ,Receptors, Immunologic ,Neoplasm Metastasis ,Aetiology ,Cancer ,Multidisciplinary ,Cultured ,Chemotaxis ,Tumor Cells ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Disease Progression ,Intercellular Signaling Peptides and Proteins ,Female ,Signal transduction ,Signal Transduction ,Endothelium ,General Science & Technology ,Breast Neoplasms ,Nerve Tissue Proteins ,Biology ,Article ,03 medical and health sciences ,Downregulation and upregulation ,Breast Cancer ,medicine ,Genetics ,Animals ,Humans ,RNA, Double-Stranded ,Animal ,Intravasation ,Endothelial Cells ,medicine.disease ,Toll-Like Receptor 3 ,Disease Models, Animal ,030104 developmental biology ,Cancer cell ,TLR3 ,Disease Models ,Cancer research ,RNA - Abstract
Blood vessels support tumours by providing nutrients and oxygen, while also acting as conduits for the dissemination of cancer(1). Here we use mouse models of breast and lung cancer to investigate whether endothelial cells also have active ‘instructive’ roles in the dissemination of cancer. We purified genetically tagged endothelial ribosomes and their associated transcripts from highly and poorly metastatic tumours. Deep sequencing revealed that metastatic tumours induced expression of the axon-guidance gene Slit2 in endothelium, establishing differential expression between the endothelial (high Slit2 expression) and tumoural (low Slit2 expression) compartments. Endothelial-derived SLIT2 protein and its receptor ROBO1 promoted the migration of cancer cells towards endothelial cells and intravasation. Deleting endothelial Slit2 suppressed metastatic dissemination in mouse models of breast and lung cancer. Conversely, deletion of tumoural Slit2 enhanced metastatic progression. We identified double-stranded RNA derived from tumour cells as an upstream signal that induces expression of endothelial SLIT2 by acting on the RNA-sensing receptor TLR3. Accordingly, a set of endogenous retroviral element RNAs were upregulated in metastatic cells and detected extracellularly. Thus, cancer cells co-opt innate RNA sensing to induce a chemotactic signalling pathway in endothelium that drives intravasation and metastasis. These findings reveal that endothelial cells have a direct instructive role in driving metastatic dissemination, and demonstrate that a single gene (Slit2) can promote or suppress cancer progression depending on its cellular source.
- Published
- 2020
91. Blocking Short-Form Ron Eliminates Breast Cancer Metastases through Accumulation of Stem-Like CD4+ T Cells That Subvert Immunosuppression
- Author
-
H. Atakan Ekiz, Amanda Jiang, Alana L. Welm, Elvelyn Fernandez, Harika Gundlapalli, and Shu-Chin Alicia Lai
- Subjects
CD4-Positive T-Lymphocytes ,T cell ,medicine.medical_treatment ,Breast Neoplasms ,Article ,Mice ,Breast cancer ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,Immunosuppression Therapy ,Lung ,business.industry ,Kinase ,Receptor Protein-Tyrosine Kinases ,Immunosuppression ,Immunotherapy ,medicine.disease ,Metastatic breast cancer ,medicine.anatomical_structure ,Oncology ,Cancer research ,Female ,business ,CD8 - Abstract
Immunotherapy has potential to prevent and treat metastatic breast cancer, but strategies to enhance immune-mediated killing of metastatic tumors are urgently needed. We report that a ligand-independent isoform of Ron kinase (SF-Ron) is a key target to enhance immune infiltration and eradicate metastatic tumors. Host-specific deletion of SF-Ron caused recruitment of lymphocytes to micrometastases, augmented tumor-specific T-cell responses, and nearly eliminated breast cancer metastasis in mice. Lack of host SF-Ron caused stem-like TCF1+ CD4+ T cells with type I differentiation potential to accumulate in metastases and prevent metastatic outgrowth. There was a corresponding increase in tumor-specific CD8+ T cells, which were also required to eliminate lung metastases. Treatment of mice with a Ron kinase inhibitor increased tumor-specific CD8+ T cells and protected from metastatic outgrowth. These data provide a strong preclinical rationale to pursue small-molecule Ron kinase inhibitors for the prevention and treatment of metastatic breast cancer. Significance: The discovery that SF-Ron promotes antitumor immune responses has significant clinical implications. Therapeutic antibodies targeting full-length Ron may not be effective for immunotherapy; poor efficacy of such antibodies in trials may be due to their inability to block SF-Ron. Our data warrant trials with inhibitors targeting SF-Ron in combination with immunotherapy. This article is highlighted in the In This Issue feature, p. 2945
- Published
- 2020
92. Ligand-based Discovery of Novel Small Molecule Inhibitors of RON Receptor Tyrosine Kinase
- Author
-
Alana L. Welm, Stéphane L Raeppel, Omid Zarei, Hariprasad Vankayalapati, Maryam Hamzeh-Mivehroud, and Najme Faham
- Subjects
Ligands ,01 natural sciences ,Receptor tyrosine kinase ,03 medical and health sciences ,Structural Biology ,Drug Discovery ,Receptor ,Glucans ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,ADME ,0303 health sciences ,Virtual screening ,biology ,Chemistry ,Organic Chemistry ,Receptor Protein-Tyrosine Kinases ,Ligand (biochemistry) ,Small molecule ,0104 chemical sciences ,Computer Science Applications ,010404 medicinal & biomolecular chemistry ,Biochemistry ,biology.protein ,Molecular Medicine ,Phosphorylation ,Signal Transduction - Abstract
Background RON (Recepteur d'Origine Nantais) receptor tyrosine kinase is a promising target for anti-cancer therapeutics. The aim of this study was to identify new RON inhibitors using virtual screening methods. Methods To this end, a ligand-based virtual screening approach was employed for screening of ZINC database on the homology model of RON receptor. All the selected hits were inspected in terms of drug-likeness, ADME properties, and toxicity profiles. Ligand-based similarity searches along with further filtering criteria led to the identification of two compounds, TKI1 and TKI2 that were evaluated using in vitro cell-based RON inhibition assays. Results The results showed that TKI1 and TKI2 could reduce phosphorylation of RON. Both compounds showed inhibitory activity of the downstream mTOR pathway with no apparent effects on other signaling mediators in a dose-dependent manner. Conclusion These compounds can provide a basis for developing novel anti-RON inhibitors applicable to cancer therapy using medicinal chemistry-oriented optimization strategies.
- Published
- 2020
93. Inhibiting an RBM39/MLL1 epigenomic regulatory complex with dominant-negative peptides disrupts cancer cell transcription and proliferation
- Author
-
Alana L. Welm, Anne M. Moon, Chad D. Huff, Yao Yu, Pavan Kumar Puvvula, Kaelan Renaldo Sullivan, Henok Eyob, and Julian Rosenberg
- Subjects
0301 basic medicine ,Epigenomics ,Transcription, Genetic ,Carcinogenesis ,Cell Survival ,Breast Neoplasms ,Cell-Penetrating Peptides ,Mice, SCID ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Splicing factor ,0302 clinical medicine ,Protein Domains ,Mice, Inbred NOD ,Cell Line, Tumor ,Gene expression ,Coactivator ,Transcriptional regulation ,Human Umbilical Vein Endothelial Cells ,Animals ,Humans ,Epigenetics ,Promoter Regions, Genetic ,Cell Proliferation ,Cell Nucleus ,Chromatin binding ,RNA-Binding Proteins ,Histone-Lysine N-Methyltransferase ,Cell biology ,Histone Code ,030104 developmental biology ,HEK293 Cells ,Phenotype ,Cancer cell ,Female ,Peptides ,030217 neurology & neurosurgery ,Myeloid-Lymphoid Leukemia Protein ,Protein Binding - Abstract
Summary RBM39 is a known splicing factor and coactivator. Here, we report that RBM39 functions as a master transcriptional regulator that interacts with the MLL1 complex to facilitate chromatin binding and H3K4 trimethylation in breast cancer cells. We identify RBM39 functional domains required for DNA and complex binding and show that the loss of RBM39 has widespread effects on H3K4me3 and gene expression, including key oncogenic pathways. RBM39’s RNA recognition motif 3 (RRM3) functions as a dominant-negative domain; namely, it disrupts the complex and H3K4me trimethylation and expression of RBM/MLL1 target genes. RRM3-derived cell-penetrating peptides phenocopy the effects of the loss of RBM39 to decrease growth and survival of all major subtypes of breast cancer and yet are nontoxic to normal cells. These findings establish RBM39/MLL1 as a major contributor to the abnormal epigenetic landscape in breast cancer and lay the foundation for peptide-mediated cancer-specific therapy based on disruption of RBM39 epigenomic functions.
- Published
- 2020
94. CD229 CAR T cells eliminate multiple myeloma and tumor propagating cells without fratricide
- Author
-
Jens Panse, Sandra D. Scherer, Rodney R. Miles, William Matsui, Michael Olson, Yasmina Noubia Abdiche, Erica R. Vander Mause, Alana L. Welm, Patricia Davis, K. David Li, Sabarinath Venniyil Radhakrishnan, Sara Yousef, Djordje Atanackovic, and Tim Luetkens
- Subjects
Male ,0301 basic medicine ,T-Lymphocytes ,Science ,medicine.medical_treatment ,Receptors, Antigen, T-Cell ,General Physics and Astronomy ,Cancer immunotherapy ,Myeloma ,Plasma cell ,Immunotherapy, Adoptive ,Article ,Antibodies ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Mice, Inbred NOD ,Signaling Lymphocytic Activation Molecule Family ,Cell surface receptor ,medicine ,Animals ,Humans ,lcsh:Science ,Receptor ,Multiple myeloma ,B-Lymphocytes ,Cell therapies ,Multidisciplinary ,Chemistry ,General Chemistry ,Immunotherapy ,medicine.disease ,Xenograft Model Antitumor Assays ,Chimeric antigen receptor ,In vitro ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer research ,lcsh:Q ,K562 Cells ,Multiple Myeloma - Abstract
Multiple myeloma (MM) is a plasma cell malignancy and most patients eventually succumb to the disease. Chimeric antigen receptor (CAR) T cells targeting B-Cell Maturation Antigen (BCMA) on MM cells have shown high-response rates, but limited durability. CD229/LY9 is a cell surface receptor present on B and T lymphocytes that is universally and strongly expressed on MM plasma cells. Here, we develop CD229 CAR T cells that are highly active in vitro and in vivo against MM plasma cells, memory B cells, and MM-propagating cells. We do not observe fratricide during CD229 CAR T cell production, as CD229 is downregulated in T cells during activation. In addition, while CD229 CAR T cells target normal CD229high T cells, they spare functional CD229neg/low T cells. These findings indicate that CD229 CAR T cells may be an effective treatment for patients with MM., CD229 is expressed on the surface of multiple myeloma cells, as well as B and T lymphocytes. Here, the authors engineer CD229-specific CAR T cells and, using patient samples and mouse models, show that treatment with these cells reduces tumour burden and results in limited targeting of T cells.
- Published
- 2020
95. Abstract PD2-01: A platform of CDK4/6 inhibitor-resistant patient-derived breast cancer organoids illuminates mechanisms of resistance and therapeutic vulnerabilities
- Author
-
Ariella B. Hanker, Sumanta Chatterjee, Yunguan Wang, Dan Ye, Dhivya R. Sudhan, Brian M. Larsen, Lauren C. Smith, Yilin Zhang, Vishal Kandagatla, Kuntal Majmudar, Ezequiel Renzulli, Saurabh Mendiratta, Kimberly Blackwell, Alana L. Welm, Sunati Sahoo, Nisha Unni, Cheryl M. Lewis, Tao Wang, Ameen A. Salahudeen, and Carlos L. Arteaga
- Subjects
Cancer Research ,Oncology - Abstract
CDK4/6 inhibitors (CDK4/6i) in combination with antiestrogens have revolutionized the treatment of ER+ metastatic breast cancer (MBC), significantly prolonging survival. However, this combination is not curative, and tumors eventually acquire resistance. Following progression on this combination, patients are left with limited treatment options. A diverse array of mechanisms of resistance to CDK4/6i + antiestrogens have been described. However, laboratory models that capture this heterogeneity of resistance mechanisms are lacking. Patient-derived organoids (PDOs) provide a rapid, robust and reliable platform that recapitulates intra-tumor heterogeneity, partially mimics the cancer microenvironment, and accurately predicts drug response. We aspired to generate a platform of CDK4/6i-resistant breast cancer PDOs to serve as models for understanding acquired resistance to CDK4/6i + antiestrogens and identifying therapies to overcome resistance. We successfully established 16 PDOs out of 32 biopsies (50% efficiency) of metastates from patients with ER+ MBC progressing on CDK4/6i (palbociclib or abemaciclib) + antiestrogens (letrozole or fulvestrant; median response to combination = 9 months). Our collection includes PDOs derived from lobular (n=3) and inflammatory (n=2) breast cancers and reflects racial/ethnic diversity (50% white/not Hispanic; 18.8% Hispanic; 12.5% Black; 12.5% other/unknown). Next-gen sequencing reports were available for 10 patients from which organoids were established, revealing alterations associated with CDK4/6i and/or antiestrogen resistance, including ESR1 (n=2), HER2/ERBB2 (n=2), PTEN (n=2), CCNE1 (n=1), NF1 (n=1), and ARID1A (n=1). Furthermore, one biopsy and its derived organoid lost ER expression, and 5 harbored PIK3CA activating mutations. Thus far, we have performed targeted DNA-sequencing on 7 PDOs and found 13/15 (86.7%) concordance with driver mutations from tumor NGS reports. PDOs established from CDK4/6i-resistant biopsies maintained resistance to palbociclib or abemaciclib ± fulvestrant (500 nM each) in 3D cell viability assays (6 days of treatment). In contrast, control PDOs established from primary ER+ breast cancer surgical samples (n=2) were sensitive to each CDK4/6i ± fulvestrant (median viability for combination=25.6-31.5% for control vs 65.2-80.5% for resistant). GSEA analysis of RNA-seq data from control (n=2) and CDK4/6i-resistant (n=6) PDOs cultured in estrogen-depleted media ± 200 nM palbociclib revealed that palbociclib treatment resulted in downregulation of E2F target and G2M checkpoint signatures in control but not resistant PDOs. Next, we performed a high-throughput screen of 1,000 compounds in 3 resistant PDOs. One PDO showed exquisite sensitivity to G2/M cell cycle checkpoint components, including CDK1, PLK1, Aurora kinase, ATR, Chk1, and Wee1 inhibitors. Finally, treatment of 10 resistant PDOs with the CDK2/4/6 inhibitor PF-06873600 revealed that the CCNE1 (cyclin E1)-amplified PDO was highly sensitive (IC50=130 nM vs >1000 nM), supporting that CCNE1-amplified tumors are vulnerable to CDK2 inhibition. Conclusions: PDOs can be successfully established from ER+ MBC biopsies, maintain the resistant phenotype in culture, retain driver alterations found in tumors from which they were derived, and fail to suppress E2F targets following treatment with CDK4/6i. Therefore, these PDOs represent valuable models to understand and explore diverse mechanisms of CDK4/6i resistance and therapeutic vulnerabilities. Citation Format: Ariella B. Hanker, Sumanta Chatterjee, Yunguan Wang, Dan Ye, Dhivya R. Sudhan, Brian M. Larsen, Lauren C. Smith, Yilin Zhang, Vishal Kandagatla, Kuntal Majmudar, Ezequiel Renzulli, Saurabh Mendiratta, Kimberly Blackwell, Alana L. Welm, Sunati Sahoo, Nisha Unni, Cheryl M. Lewis, Tao Wang, Ameen A. Salahudeen, Carlos L. Arteaga. A platform of CDK4/6 inhibitor-resistant patient-derived breast cancer organoids illuminates mechanisms of resistance and therapeutic vulnerabilities [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr PD2-01.
- Published
- 2022
96. Understanding the Bone in Cancer Metastasis
- Author
-
Jaime Fornetti, Sheila A Stewart, and Alana L. Welm
- Subjects
0301 basic medicine ,Aging ,Endocrinology, Diabetes and Metabolism ,Cell Communication ,Bone and Bones ,Metastasis ,Bone remodeling ,03 medical and health sciences ,Breast cancer ,Prostate ,Tumor Microenvironment ,medicine ,Animals ,Homeostasis ,Humans ,Orthopedics and Sports Medicine ,Neoplasm Metastasis ,Tumor microenvironment ,business.industry ,Melanoma ,Cancer ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Cancer cell ,Cancer research ,business - Abstract
The bone is the third most common site of metastasis for a wide range of solid tumors including lung, breast, prostate, colorectal, thyroid, gynecologic, and melanoma, with 70% of metastatic prostate and breast cancer patients harboring bone metastasis.1 Unfortunately, once cancer spreads to the bone, it is rarely cured and is associated with a wide range of morbidities including pain, increased risk of fracture, and hypercalcemia. This fact has driven experts in the fields of bone and cancer biology to study the bone, and has revealed that there is a great deal that each can teach the other. The complexity of the bone was first described in 1889 when Stephen Paget proposed that tumor cells have a proclivity for certain organs, where they "seed" into a friendly "soil" and eventually grow into metastatic lesions. Dr. Paget went on to argue that although many study the "seed" it would be paramount to understand the "soil." Since this original work, significant advances have been made not only in understanding the cell-autonomous mechanisms that drive metastasis, but also alterations which drive changes to the "soil" that allow a tumor cell to thrive. Indeed, it is now clear that the "soil" in different metastatic sites is unique, and thus the mechanisms that allow tumor cells to remain in a dormant or growing state are specific to the organ in question. In the bone, our knowledge of the components that contribute to this fertile "soil" continues to expand, but our understanding of how they impact tumor growth in the bone remains in its infancy. Indeed, we now appreciate that the endosteal niche likely contributes to tumor cell dormancy, and that osteoclasts, osteocytes, and adipocytes can impact tumor cell growth. Here, we discuss the bone microenvironment and how it impacts cancer cell seeding, dormancy, and growth. © 2018 American Society for Bone and Mineral Research.
- Published
- 2018
97. EPHB6 augments both development and drug sensitivity of triple-negative breast cancer tumours
- Author
-
Amanda Gall, C. Ronald Geyer, Deborah H. Anderson, John F. DeCoteau, Mohsin Ali, Morgan W. B. Kirzinger, Anthony Kusalik, Amr M. El Zawily, Andrew Freywald, TaeHyung Kim, Alana L. Welm, Odette Allonby, Franco J. Vizeacoumar, Matthew Shannon, Behzad M. Toosi, Tanya Freywald, Mohan Babu, Darrell D. Mousseau, Frederick S. Vizeacoumar, Peter M. Siegel, and Luke Truitt
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Cancer Research ,Epithelial-Mesenchymal Transition ,MAP Kinase Signaling System ,Receptor, ErbB-2 ,Mice, Nude ,Triple Negative Breast Neoplasms ,Biology ,Article ,Receptor tyrosine kinase ,Mice ,03 medical and health sciences ,Breast cancer ,Cell Line, Tumor ,Genetics ,medicine ,EPHB6 ,Animals ,Humans ,Receptor ,Molecular Biology ,Transcription factor ,Triple-negative breast cancer ,Cell Proliferation ,Receptors, Eph Family ,Erythropoietin-producing hepatocellular (Eph) receptor ,medicine.disease ,030104 developmental biology ,Drug Resistance, Neoplasm ,Neoplastic Stem Cells ,ras Proteins ,Cancer research ,biology.protein ,Female ,Neoplasm Recurrence, Local ,Octamer Transcription Factor-3 ,DNA Damage - Abstract
Triple-negative breast cancer (TNBC) tumours that lack expression of oestrogen, and progesterone receptors, and do not overexpress the HER2 receptor represent the most aggressive breast cancer subtype, which is characterised by the resistance to therapy in frequently relapsing tumours and a high rate of patient mortality. This is likely due to the resistance of slowly proliferating tumour-initiating cells (TICs), and understanding molecular mechanisms that control TICs behaviour is crucial for the development of effective therapeutic approaches. Here, we present our novel findings, indicating that an intrinsically catalytically inactive member of the Eph group of receptor tyrosine kinases, EPHB6, partially suppresses the epithelial–mesenchymal transition in TNBC cells, while also promoting expansion of TICs. Our work reveals that EPHB6 interacts with the GRB2 adapter protein and that its effect on enhancing cell proliferation is mediated by the activation of the RAS-ERK pathway, which allows it to elevate the expression of the TIC-related transcription factor, OCT4. Consistent with this, suppression of either ERK or OCT4 activities blocks EPHB6-induced pro-proliferative responses. In line with its ability to trigger propagation of TICs, EPHB6 accelerates tumour growth, potentiates tumour initiation and increases TIC populations in xenograft models of TNBC. Remarkably, EPHB6 also suppresses tumour drug resistance to DNA-damaging therapy, probably by forcing TICs into a more proliferative, drug-sensitive state. In agreement, patients with higher EPHB6 expression in their tumours have a better chance for recurrence-free survival. These observations describe an entirely new mechanism that governs TNBC and suggest that it may be beneficial to enhance EPHB6 action concurrent with applying a conventional DNA-damaging treatment, as it would decrease drug resistance and improve tumour elimination.
- Published
- 2018
98. CD229 CAR T Cell Therapy for the Treatment of Relapsed B Cell Lymphoma
- Author
-
Djordje Atanackovic, Jennie Y. Law, Nicola J. Camp, Michael L. Olson, Stephanie Avila, Alana L. Welm, Aaron P. Rapoport, Erica Vander Mause, Tim Luetkens, Fiorella Iglesias, Saurabh Dahiya, Sabarinath Venniyil Radhakrishnan, Erin Morales, and Joshua Brody
- Subjects
immune system diseases ,business.industry ,hemic and lymphatic diseases ,Immunology ,Cancer research ,medicine ,CAR T-cell therapy ,Cell Biology ,Hematology ,B-cell lymphoma ,medicine.disease ,business ,Biochemistry - Abstract
B cell lymphoma is the most common hematologic malignancy in the United States. Although treatment options have greatly improved in the past several decades, outcomes for patients with relapsed B cell lymphoma remain poor. Chimeric antigen receptor (CAR) T cells have recently entered the clinic with promise to address the gap in effective therapies for patients relapsed B cell lymphoma. However, antigen loss and poor CAR T cell persistence has been shown to drive resistance to the widely approved CD19-targeted CAR in some patients, demonstrating the need for additional therapies. Here, we demonstrate CD229-targeted CAR T cell therapy as a promising option for the treatment of relapsed B cell lymphoma, addressing an important group of patients with typically poor outcomes. CD229 is an immune-modulating receptor expressed on the surface of B cells that we recently found to be highly expressed in the plasma cell neoplasm multiple myeloma (Radhakrishnan et al. 2020). We utilized semi-quantitative PCR and flow cytometry to assess whether CD229 is also expressed on malignant B cells earlier in development as found in B cell lymphoma. Expression analysis revealed the presence of CD229 in a panel of 11 B cell lymphoma cell lines and 45 primary B cell lymphoma samples comprising several subsets of disease including aggressive B cell lymphomas such as diffuse large B cell lymphoma (DLBCL), mantle cell lymphoma (MCL) and Burkitt lymphoma as well as indolent subtypes of B cell lymphoma including chronic lymphoblastic leukemia (CLL) and follicular lymphoma. Of note, CD229 was found to be overexpressed on primary B cell lymphoma cells when compared to autologous normal B cells. Given the high levels of CD229 expression throughout all B cell lymphoma subtypes analyzed, we generated CD229 CAR T cells in order to determine whether CAR T cell therapy is an effective way to target CD229 expressing B cell lymphoma cells. CD229 CAR T cells exhibited robust cytotoxicity when cocultured with B cell lymphoma cell lines and primary samples characterized by significant production of TH1 cytokines IL-2, TNF and IFNγ and rapid loss of B cell lymphoma cell viability when compared to control CAR T cells lacking an antigen binding scFv domain (∆scFv CAR T cells). In vivo analysis revealed effective tumor control in NSG mice carrying B cell lymphoma cell lines JeKo-1 (MCL) and DB (DLBCL) when treated with CD229 CAR T cells versus ∆scFv CAR T cells. Finally, we sought to determine the efficacy of CD229 CAR T cells in the context of CD19 CAR T cell therapy relapse. Here, a 71-year-old patient with CLL had an initial response when treated with CD19 CAR T cells but quickly relapsed only 2 months after treatment. Malignant cells from the CLL patient retained CD229 expression as identified by flow cytometry and an ex vivo coculture with CD229 CAR T cells revealed robust killing of CLL cells by CD229 CAR T cells. Transfer of antigen from target cell to CAR T cell by trogocytosis was recently suggested to drive relapse following CAR T cell therapy by decreasing antigen on tumor cells and promoting CAR T cell fratricide (Hamieh et al. 2019). We cocultured CD19 and CD229 CAR T cells with primary CLL cells and assessed CD19 and CD229 expression as well as CAR T cell viability by flow cytometry. In contrast with CD19 CAR T cells, CD229 CARs did not strip their target antigen from the surface of CLL cells. The transfer of CD19 from CLL cells to CD19 CAR T cells resulted in poor CAR T cell viability while CD229 CAR T cell viability remained high following coculture. In summary, we demonstrate that CD229 is a promising therapeutic target in B cell lymphoma due to its high levels of expression throughout many subtypes of disease. CD229 CAR T cells effectively kill B cell lymphoma cells in vitro and control growth of aggressive B cell lymphomas in vivo. Finally, CD229 CAR T cells are effective against primary CLL cells from patients that have relapsed from CD19 CAR T cell therapy and do no exhibit antigen loss by trogocytosis. Taken together, these data suggest that CD229 CAR T cell therapy may be a promising option to address the poor outcomes for patients with relapsed B cell lymphoma. Disclosures No relevant conflicts of interest to declare.
- Published
- 2021
99. Abstract SY16-02: Using patient-derived models for functional precision oncology in advanced breast cancer
- Author
-
Alana L. Welm
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Advanced breast ,Cancer ,medicine.disease ,Slow growth ,Patient care ,Breast cancer ,Precision oncology ,Internal medicine ,medicine ,business - Abstract
Patient derived xenografts (PDX) are valuable, clinically-relevant models of cancer. Their close genomic, phenotypic, and temporal association with patient tumors makes them well-suited for pre-clinical and co-clinical studies that assess the potential of new therapeutics. However, PDX models are not amenable to large-scale drug sensitivity studies or real-time precision oncology due to their relatively slow growth, high cost, and low throughput. To address this, we established and characterized organoid lines from breast cancer patient tumors and PDXs and evaluated their fidelity to the original tumors and their utility in high-throughput therapeutic studies. Results of comparisons between in vitro and in vivo patient-derived models of breast cancer will be presented, along with extension of this technology to inform patient care in an IRB-approved study of precision oncology. Our work demonstrates that breast cancer organoid models are a powerful parallel resource to PDX models, especially useful for efficient determination of drug responses, and are amenable to informing real-time patient care in a functional precision oncology setting. Citation Format: Alana L. Welm. Using patient-derived models for functional precision oncology in advanced breast cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr SY16-02.
- Published
- 2021
100. PDX-MI: Minimal Information for Patient-Derived Tumor Xenograft Models
- Author
-
Matthew H. Brush, Theodore C. Goldstein, Yvonne A. Evrard, Nathalie Conte, Melissa A. Haendel, Kevin C K Lloyd, Annette T. Byrne, Peter J. Houghton, Carlos Caldas, Amanda L. Christie, Frédéric Amant, Alana L. Welm, Stefan M. Pfister, Mark A. Murakami, Jos Jonkers, Patrick Dunn, Tin Oo Khor, Danielle Greenawalt, Jonathan R. Dry, David M. Weinstock, Sebastian Brabetz, Oscar M. Rueda, Zhiping Gu, Giorgio Inghirami, Dominic Clark, Olivier Duchamp, James M. Olson, Emilie Vinolo, Neal Goodwin, Kristopher K. Frese, Robert J. Wechsler-Reya, Terrence F. Meehan, Daniel S. Peeper, Marcel Kool, Enzo Medico, Jeremy Mason, Stephane Ferretti, Carol J. Bult, Atul J. Butte, S. John Weroha, Els Hermans, Kristel Kemper, Alejandra Bruna, Heidi Dowst, Je Kyung Seong, James H. Doroshow, Livio Trusolino, Michael T. Lewis, Jeffrey Wiser, Dale A. Begley, Steven B. Neuhauser, Helen Parkinson, Debra M. Krupke, Andrea Bertotti, Other departments, Obstetrics and Gynaecology, and ARD - Amsterdam Reproduction and Development
- Subjects
0301 basic medicine ,Oncology ,endocrine system ,Cancer Research ,medicine.medical_specialty ,Patients ,endocrine system diseases ,Oncology and Carcinogenesis ,Bioinformatics ,digestive system ,Article ,Mice ,03 medical and health sciences ,Neoplasms ,Internal medicine ,medicine ,Drug response ,Animals ,Humans ,Oncology & Carcinogenesis ,Tumor xenograft ,Cancer ,Databases as Topic ,Disease Models, Animal ,Xenograft Model Antitumor Assays ,Mouse strain ,Animal ,Extramural ,business.industry ,nutritional and metabolic diseases ,medicine.disease ,Good Health and Well Being ,030104 developmental biology ,Disease Models ,Research studies ,business ,hormones, hormone substitutes, and hormone antagonists - Abstract
Patient-derived tumor xenograft (PDX) mouse models have emerged as an important oncology research platform to study tumor evolution, mechanisms of drug response and resistance, and tailoring chemotherapeutic approaches for individual patients. The lack of robust standards for reporting on PDX models has hampered the ability of researchers to find relevant PDX models and associated data. Here we present the PDX models minimal information standard (PDX-MI) for reporting on the generation, quality assurance, and use of PDX models. PDX-MI defines the minimal information for describing the clinical attributes of a patient's tumor, the processes of implantation and passaging of tumors in a host mouse strain, quality assurance methods, and the use of PDX models in cancer research. Adherence to PDX-MI standards will facilitate accurate search results for oncology models and their associated data across distributed repository databases and promote reproducibility in research studies using these models. Cancer Res; 77(21); e62–66. ©2017 AACR.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.