44 results on '"Zabransky DJ"'
Search Results
2. Abstract PD2-05: Inhibition of mutant HER2 results in synthetic lethality when combined with ER antagonists in ER+/HER2 mutant human breast cancer cells
- Author
-
Croessmann, S, primary, Zabransky, DJ, additional, Cutler, RE, additional, Lalani, AS, additional, Park, BH, additional, and Arteaga, CL, additional
- Published
- 2017
- Full Text
- View/download PDF
3. Author Correction: Stromal changes in the aged lung induce an emergence from melanoma dormancy.
- Author
-
Fane ME, Chhabra Y, Alicea GM, Maranto DA, Douglass SM, Webster MR, Rebecca VW, Marino GE, Almeida F, Ecker BL, Zabransky DJ, Hüser L, Beer T, Tang HY, Kossenkov A, Herlyn M, Speicher DW, Xu W, Xu X, Jaffee EM, Aguirre-Ghiso JA, and Weeraratna AT
- Published
- 2025
- Full Text
- View/download PDF
4. Cancer associated fibroblasts drive epithelial to mesenchymal transition and classical to basal change in pancreatic ductal adenocarcinoma cells with loss of IL-8 expression.
- Author
-
Guinn S, Perez B, Tandurella JA, Ramani M, Lee JW, Zabransky DJ, Kartalia E, Patel J, Zlomke H, Nicolson N, Shin S, Barrett B, Sun N, Hernandez A, Coyne E, Cannon C, Gross NE, Charmsaz S, Cho Y, Leatherman J, Lyman M, Mitchell J, Kagohara LT, Goggins MG, Lafaro KJ, He J, Shubert C, Burns W, Zheng L, Fertig EJ, Jaffee EM, Burkhart RA, Ho WJ, and Zimmerman JW
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) carries an extremely poor prognosis, in part resulting from cellular heterogeneity that supports overall tumorigenicity. Cancer associated fibroblasts (CAF) are key determinants of PDAC biology and response to systemic therapy. While CAF subtypes have been defined, the effects of patient-specific CAF heterogeneity and plasticity on tumor cell behavior remain unclear. Here, multi-omics was used to characterize the tumor microenvironment (TME) in tumors from patients undergoing curative-intent surgery for PDAC. In these same patients, matched tumor organoid and CAF lines were established to functionally validate the impact of CAFs on the tumor cells. CAFs were found to drive epithelial-mesenchymal transition (EMT) and a switch in tumor cell classificiaton from classical to basal subtype. Furthermore, we identified CAF-specific interleukin 8 (IL-8) as an important modulator of tumor cell subtype. Finally, we defined neighborhood relationships between tumor cell and T cell subsets., Competing Interests: Conflict of Interest Statement E.M.J is a paid consultant for Adaptive Biotech, Achilles, DragonFly, Candel Therapeutics, Genocea, and Roche. She receives funding from Lustgarten Foundation and Bristol Myer Squibb. She is the Chief Medical Advisor for Lustgarten and SAB advisor to the Parker Institute for Cancer Immunotherapy (PICI) and for the C3 Cancer Institute. She is a founding member of Abmeta. E.J.F is on the SAB for Resistance Biology, Consultant for Mestag Therapeutics and Merck. L.Z. reports personal fees from Biosion, Alphamab, NovaRock, Xilio, Ambrx, Novagenesis, and Snow Lake Capitals; and other support from Alphamab and Mingruizhiyao outside the submitted work. D.J.Z. reports grant support (to Johns Hopkins) and travel from Roche/Genentech, and honoraria from Omni Health Media, Sermo, Atheneum, Escientiq, Deerfield Institute, ZoomRx, and NeuCore Bio.W.J.H. reports patent royalties from Rodeo/Amgen; grants from Sanofi, NeoTX, and Riboscience; speaking/travel honoraria from Exelixis and Standard BioTools. J.W.Z. report grant funding support (to Johns Hopkins) and travel from Roche/Genentch outside the submitted work.
- Published
- 2025
- Full Text
- View/download PDF
5. Tumor-derived CCL2 drives tumor growth and immunosuppression in IDH1- mutant cholangiocarcinoma.
- Author
-
Zabransky DJ, Kartalia E, Lee JW, Leatherman JM, Charmsaz S, Young SE, Chhabra Y, Franch-Expósito S, Kang M, Maru S, Rastkari N, Davis M, Dalton WB, Oshima K, Baretti M, Azad NS, Jaffee EM, and Yarchoan M
- Abstract
Background and Aims: Isocitrate dehydrogenase 1 ( IDH1 )-mutant cholangiocarcinoma (CCA) is a highly lethal subtype of hepatobiliary cancer that is often resistant to immune checkpoint inhibitor therapies. We evaluated the effects of IDH1 mutations in CCA cells on the tumor immune microenvironment and identified opportunities for therapeutic intervention., Approach and Results: Analysis of 2606 human CCA tumors using deconvolution of RNA-sequencing data identified decreased CD8+ T cell and increased M2-like tumor-associated macrophage (TAM) infiltration in IDH1 -mutant compared to IDH1 wild-type tumors. To model the tumor immune microenvironment of IDH1 -mutant CCA in vivo, we generated an isogenic cell line panel of mouse SB1 CCA cells containing a heterozygous IDH1 R132C (SB1 mIDH1 ) or control (SB1 WT ) mutation using CRISPR-mediated homology-directed repair. SB1 mIDH1 cells recapitulated features of human IDH1 -mutant CCA including D-2-hydroxyglutarate production and increased M2-like TAM infiltration. SB1 mIDH1 cells and tumors produced increased levels of CCL2, a chemokine involved in the recruitment and polarization of M2-like TAMs, compared to wild-type controls. In vivo neutralization of CCL2 led to decreased M2-like TAM infiltration, reduced tumor size, and improved overall survival in mice harboring SB1 mIDH1 tumors., Conclusions: IDH1- mutant CCA is characterized by an increased abundance of M2-like TAMs. Targeting CCL2 remodels the tumor immune microenvironment and improves outcomes in preclinical models of IDH1 -mutant CCA, highlighting the role of myeloid-targeted immunotherapies in the treatment of this cancer., (Copyright © 2024 American Association for the Study of Liver Diseases.)
- Published
- 2024
- Full Text
- View/download PDF
6. Immunotherapy response induces divergent tertiary lymphoid structure morphologies in hepatocellular carcinoma.
- Author
-
Shu DH, Ho WJ, Kagohara LT, Girgis A, Shin SM, Danilova L, Lee JW, Sidiropoulos DN, Mitchell S, Munjal K, Howe K, Bendinelli KJ, Kartalia E, Qi H, Mo G, Montagne J, Leatherman JM, Lopez-Vidal TY, Zhu Q, Huff AL, Yuan X, Hernandez A, Coyne EM, Zaidi N, Zabransky DJ, Engle LL, Ogurtsova A, Baretti M, Laheru D, Durham JN, Wang H, Sunshine JC, Johnston RJ, Deutsch JS, Taube JM, Anders RA, Jaffee EM, Fertig EJ, and Yarchoan M
- Subjects
- Humans, Male, Female, Middle Aged, Prognosis, Aged, Neoadjuvant Therapy methods, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Immunologic Memory, Dendritic Cells immunology, Immune Checkpoint Inhibitors therapeutic use, Immune Checkpoint Inhibitors pharmacology, Carcinoma, Hepatocellular immunology, Carcinoma, Hepatocellular therapy, Carcinoma, Hepatocellular pathology, Liver Neoplasms immunology, Liver Neoplasms therapy, Liver Neoplasms pathology, Tertiary Lymphoid Structures immunology, Tertiary Lymphoid Structures pathology, Immunotherapy methods
- Abstract
Tertiary lymphoid structures (TLS) are associated with improved response in solid tumors treated with immune checkpoint blockade, but understanding of the prognostic and predictive value of TLS and the circumstances of their resolution is incomplete. Here we show that in hepatocellular carcinoma treated with neoadjuvant immunotherapy, high intratumoral TLS density at the time of surgery is associated with pathologic response and improved relapse-free survival. In areas of tumor regression, we identify a noncanonical involuted morphology of TLS marked by dispersion of the B cell follicle, persistence of a T cell zone enriched for T cell-mature dendritic cell interactions and increased expression of T cell memory markers. Collectively, these data suggest that TLS can serve as both a prognostic and predictive marker of response to immunotherapy in hepatocellular carcinoma and that late-stage TLS may support T cell memory formation after elimination of a viable tumor., (© 2024. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2024
- Full Text
- View/download PDF
7. Sex-dependent effects in the aged melanoma tumor microenvironment influence invasion and resistance to targeted therapy.
- Author
-
Chhabra Y, Fane ME, Pramod S, Hüser L, Zabransky DJ, Wang V, Dixit A, Zhao R, Kumah E, Brezka ML, Truskowski K, Nandi A, Marino-Bravante GE, Carey AE, Gour N, Maranto DA, Rocha MR, Harper EI, Ruiz J, Lipson EJ, Jaffee EM, Bibee K, Sunshine JC, Ji H, and Weeraratna AT
- Subjects
- Animals, Male, Mice, Female, Humans, Cell Line, Tumor, Proto-Oncogene Proteins B-raf metabolism, Proto-Oncogene Proteins B-raf genetics, Skin Neoplasms pathology, Skin Neoplasms drug therapy, Skin Neoplasms metabolism, Fibroblasts metabolism, Neoplasm Invasiveness, Axl Receptor Tyrosine Kinase, Receptor Protein-Tyrosine Kinases metabolism, Proto-Oncogene Proteins metabolism, Cellular Senescence, Sex Characteristics, Cell Proliferation, Aging, Mice, Inbred C57BL, Tumor Microenvironment, Melanoma pathology, Melanoma drug therapy, Melanoma metabolism, Drug Resistance, Neoplasm, Bone Morphogenetic Protein 2 metabolism, Enhancer of Zeste Homolog 2 Protein metabolism
- Abstract
There is documented sex disparity in cutaneous melanoma incidence and mortality, increasing disproportionately with age and in the male sex. However, the underlying mechanisms remain unclear. While biological sex differences and inherent immune response variability have been assessed in tumor cells, the role of the tumor-surrounding microenvironment, contextually in aging, has been overlooked. Here, we show that skin fibroblasts undergo age-mediated, sex-dependent changes in their proliferation, senescence, ROS levels, and stress response. We find that aged male fibroblasts selectively drive an invasive, therapy-resistant phenotype in melanoma cells and promote metastasis in aged male mice by increasing AXL expression. Intrinsic aging in male fibroblasts mediated by EZH2 decline increases BMP2 secretion, which in turn drives the slower-cycling, highly invasive, and therapy-resistant melanoma cell phenotype, characteristic of the aged male TME. Inhibition of BMP2 activity blocks the emergence of invasive phenotypes and sensitizes melanoma cells to BRAF/MEK inhibition., Competing Interests: Declaration of interests A.T.W. is on the board of reGAIN Therapeutics and the Melanoma Research Foundation. E.M.J. reports other support from Abmeta, personal fees from Genocea, personal fees from Achilles, personal fees from DragonFly, personal fees from Candel Therapeutics, other support from the Parker Institute, grants and other support from Lustgarten, personal fees from Carta, grants and other support from Genentech, grants and other support from AstraZeneca, personal fees from NextCure, and grants and other support from Break Through Cancer outside of the submitted work. D.J.Z. reports grant funding (paid to Johns Hopkins University) from Roche/Genentech. Y.C. and M.E.F. are affiliated with the Cancer Signaling and Microenvironment program, FoxChase Cancer Center, Philadelphia, PA, USA., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. Aged fibroblast-derived extracellular vesicles promote angiogenesis in melanoma.
- Author
-
Hüser L, Chhabra Y, Gololobova O, Wang V, Liu G, Dixit A, Rocha MR, Harper EI, Fane ME, Marino-Bravante GE, Zabransky DJ, Cai KQ, Utikal J, Slusher BS, Walston J, Lipson EJ, Witwer KW, and Weeraratna AT
- Subjects
- Humans, Tumor Microenvironment, Cell Line, Tumor, Skin Neoplasms pathology, Skin Neoplasms metabolism, Skin Neoplasms genetics, Aging metabolism, Aging pathology, Endothelial Cells metabolism, Endothelial Cells pathology, Animals, Angiogenesis, Extracellular Vesicles metabolism, Melanoma metabolism, Melanoma pathology, Neovascularization, Pathologic metabolism, Neovascularization, Pathologic pathology, Fibroblasts metabolism, Fibroblasts pathology, Tetraspanin 29 metabolism, Tetraspanin 29 genetics
- Abstract
Advancing age is a negative prognostic factor for cutaneous melanoma. However, the role of extracellular vesicles (EVs) within the melanoma tumor microenvironment (TME) has remained unexplored in the context of aging. While the size and morphology of the EVs isolated from young vs. aged fibroblasts remained unaltered, the contents of the protein cargo were changed. Aging reduced the expression of the tetraspanin CD9 in both the dermal fibroblasts and released EVs. CD9 is a crucial regulator of EV cargo sorting. Modulating the CD9 expression in fibroblasts was sufficient to alter its levels in EVs. Mass spectrometry analysis of EVs released by CD9 knockdown (KD) vs. control cells revealed a significant increase in angiopoietin-like protein 2 (ANGPTL2), an angiogenesis promoter. Analysis of primary endothelial cells confirmed increased sprouting under CD9 KD conditions. Together, our data indicate that aged EVs play an important role in promoting a tumor-permissive microenvironment., Competing Interests: Declaration of interests A.T.W. is on the boards of reGAIN Therapeutics and the Melanoma Research Foundation and the scientific advisory committee of the V Foundation. D.J.Z. reports grant funding (paid to Johns Hopkins University) from Roche/Genentech., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
9. Transfer Learning Reveals Cancer-Associated Fibroblasts Are Associated with Epithelial-Mesenchymal Transition and Inflammation in Cancer Cells in Pancreatic Ductal Adenocarcinoma.
- Author
-
Guinn S, Kinny-Köster B, Tandurella JA, Mitchell JT, Sidiropoulos DN, Loth M, Lyman MR, Pucsek AB, Zabransky DJ, Lee JW, Kartalia E, Ramani M, Seppälä TT, Cherry C, Suri R, Zlomke H, Patel J, He J, Wolfgang CL, Yu J, Zheng L, Ryan DP, Ting DT, Kimmelman A, Gupta A, Danilova L, Elisseeff JH, Wood LD, Stein-O'Brien G, Kagohara LT, Jaffee EM, Burkhart RA, Fertig EJ, and Zimmerman JW
- Subjects
- Humans, Organoids pathology, Organoids metabolism, Vascular Endothelial Growth Factor A metabolism, Vascular Endothelial Growth Factor A genetics, Neuropilin-1 metabolism, Neuropilin-1 genetics, Gene Expression Regulation, Neoplastic, Cell Line, Tumor, Cell Communication, Epithelial-Mesenchymal Transition, Carcinoma, Pancreatic Ductal pathology, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal genetics, Cancer-Associated Fibroblasts metabolism, Cancer-Associated Fibroblasts pathology, Pancreatic Neoplasms pathology, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms genetics, Inflammation pathology, Inflammation metabolism, Tumor Microenvironment, Integrin beta1 metabolism, Integrin beta1 genetics, Coculture Techniques, Single-Cell Analysis
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy characterized by an immunosuppressive tumor microenvironment enriched with cancer-associated fibroblasts (CAF). This study used a convergence approach to identify tumor cell and CAF interactions through the integration of single-cell data from human tumors with human organoid coculture experiments. Analysis of a comprehensive atlas of PDAC single-cell RNA sequencing data indicated that CAF density is associated with increased inflammation and epithelial-mesenchymal transition (EMT) in epithelial cells. Transfer learning using transcriptional data from patient-derived organoid and CAF cocultures provided in silico validation of CAF induction of inflammatory and EMT epithelial cell states. Further experimental validation in cocultures demonstrated integrin beta 1 (ITGB1) and vascular endothelial factor A (VEGFA) interactions with neuropilin-1 mediating CAF-epithelial cell cross-talk. Together, this study introduces transfer learning from human single-cell data to organoid coculture analyses for experimental validation of discoveries of cell-cell cross-talk and identifies fibroblast-mediated regulation of EMT and inflammation., Significance: Adaptation of transfer learning to relate human single-cell RNA sequencing data to organoid-CAF cocultures facilitates discovery of human pancreatic cancer intercellular interactions and uncovers cross-talk between CAFs and tumor cells through VEGFA and ITGB1., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
10. Oncogene-induced matrix reorganization controls CD8+ T cell function in the soft-tissue sarcoma microenvironment.
- Author
-
Fuller AM, Pruitt HC, Liu Y, Irizarry-Negron VM, Pan H, Song H, DeVine A, Katti RS, Devalaraja S, Ciotti GE, Gonzalez MV, Williams EF, Murazzi I, Ntekoumes D, Skuli N, Hakonarson H, Zabransky DJ, Trevino JG, Weeraratna A, Weber K, Haldar M, Fraietta JA, Gerecht S, and Eisinger-Mathason TSK
- Subjects
- Animals, Mice, Humans, Collagen Type VI genetics, Collagen Type VI immunology, Collagen Type VI metabolism, Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Adaptor Proteins, Signal Transducing immunology, Transcription Factors genetics, Transcription Factors metabolism, Transcription Factors immunology, Oncogenes, Neoplasm Proteins immunology, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Collagen Type I metabolism, Collagen Type I genetics, Collagen Type I immunology, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes pathology, Tumor Microenvironment immunology, YAP-Signaling Proteins immunology, YAP-Signaling Proteins genetics, Extracellular Matrix immunology, Extracellular Matrix metabolism, Extracellular Matrix pathology, Sarcoma immunology, Sarcoma pathology, Sarcoma genetics, Sarcoma metabolism
- Abstract
CD8+ T cell dysfunction impedes antitumor immunity in solid cancers, but the underlying mechanisms are diverse and poorly understood. Extracellular matrix (ECM) composition has been linked to impaired T cell migration and enhanced tumor progression; however, impacts of individual ECM molecules on T cell function in the tumor microenvironment (TME) are only beginning to be elucidated. Upstream regulators of aberrant ECM deposition and organization in solid tumors are equally ill-defined. Therefore, we investigated how ECM composition modulates CD8+ T cell function in undifferentiated pleomorphic sarcoma (UPS), an immunologically active desmoplastic tumor. Using an autochthonous murine model of UPS and data from multiple human patient cohorts, we discovered a multifaceted mechanism wherein the transcriptional coactivator YAP1 promotes collagen VI (COLVI) deposition in the UPS TME. In turn, COLVI induces CD8+ T cell dysfunction and immune evasion by remodeling fibrillar collagen and inhibiting T cell autophagic flux. Unexpectedly, collagen I (COLI) opposed COLVI in this setting, promoting CD8+ T cell function and acting as a tumor suppressor. Thus, CD8+ T cell responses in sarcoma depend on oncogene-mediated ECM composition and remodeling.
- Published
- 2024
- Full Text
- View/download PDF
11. Fibroblasts in the Aged Pancreas Drive Pancreatic Cancer Progression.
- Author
-
Zabransky DJ, Chhabra Y, Fane ME, Kartalia E, Leatherman JM, Hüser L, Zimmerman JW, Delitto D, Han S, Armstrong TD, Charmsaz S, Guinn S, Pramod S, Thompson ED, Hughes SJ, O'Connell J, Egan JM, Jaffee EM, and Weeraratna AT
- Subjects
- Animals, Mice, Growth Differentiation Factor 15 genetics, Growth Differentiation Factor 15 therapeutic use, Proto-Oncogene Proteins c-akt, Pancreas pathology, Fibroblasts pathology, Tumor Microenvironment, Cell Line, Tumor, Pancreatic Neoplasms pathology, Cancer-Associated Fibroblasts pathology
- Abstract
Pancreatic cancer is more prevalent in older individuals and often carries a poorer prognosis for them. The relationship between the microenvironment and pancreatic cancer is multifactorial, and age-related changes in nonmalignant cells in the tumor microenvironment may play a key role in promoting cancer aggressiveness. Because fibroblasts have profound impacts on pancreatic cancer progression, we investigated whether age-related changes in pancreatic fibroblasts influence cancer growth and metastasis. Proteomics analysis revealed that aged fibroblasts secrete different factors than young fibroblasts, including increased growth/differentiation factor 15 (GDF-15). Treating young mice with GDF-15 enhanced tumor growth, whereas aged GDF-15 knockout mice showed reduced tumor growth. GDF-15 activated AKT, rendering tumors sensitive to AKT inhibition in an aged but not young microenvironment. These data provide evidence for how aging alters pancreatic fibroblasts and promotes tumor progression, providing potential therapeutic targets and avenues for studying pancreatic cancer while accounting for the effects of aging., Significance: Aged pancreatic fibroblasts secrete GDF-15 and activate AKT signaling to promote pancreatic cancer growth, highlighting the critical role of aging-mediated changes in the pancreatic cancer microenvironment in driving tumor progression. See related commentary by Isaacson et al., p. 1185., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
12. Reply: The Hepa 1-6 may not be suitable for use in hepatocellular carcinoma models to explore responses to drug therapy.
- Author
-
Zabransky DJ, Yarchoan M, and Ho WJ
- Subjects
- Humans, Cell Line, Tumor, Carcinoma, Hepatocellular drug therapy, Carcinoma, Hepatocellular pathology, Liver Neoplasms drug therapy, Liver Neoplasms pathology
- Published
- 2024
- Full Text
- View/download PDF
13. ERK hyperactivation serves as a unified mechanism of escape in intrinsic and acquired CDK4/6 inhibitor resistance in acral lentiginous melanoma.
- Author
-
Jagirdar K, Portuallo ME, Wei M, Wilhide M, Bravo Narula JA, Robertson BM, Alicea GM, Aguh C, Xiao M, Godok T, Fingerman D, Brown GS, Herlyn M, Elad VM, Guo X, Toska E, Zabransky DJ, Wubbenhorst B, Nathanson KL, Kwatra S, Goyal Y, Ji H, Liu Q, and Rebecca VW
- Subjects
- Animals, Humans, Cyclin-Dependent Kinase 4, Cyclin-Dependent Kinase 6, Disease Models, Animal, Cell Cycle, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Melanoma drug therapy, Melanoma genetics, Melanoma pathology, Skin Neoplasms drug therapy, Skin Neoplasms genetics
- Abstract
Patients with metastatic acral lentiginous melanoma (ALM) suffer worse outcomes relative to patients with other forms of cutaneous melanoma (CM), and do not benefit as well to approved melanoma therapies. Identification of cyclin-dependent kinase 4 and 6 (CDK4/6) pathway gene alterations in >60% of ALMs has led to clinical trials of the CDK4/6 inhibitor (CDK4i/6i) palbociclib for ALM; however, median progression free survival with CDK4i/6i treatment was only 2.2 months, suggesting existence of resistance mechanisms. Therapy resistance in ALM remains poorly understood; here we report hyperactivation of MAPK signaling and elevated cyclin D1 expression serve as a mechanism of intrinsic early/adaptive CDK4i/6i resistance. ALM cells that have acquired CDK4i/6i resistance following chronic treatment exposure also exhibit hyperactivation of the MAPK pathway. MEK and/or ERK inhibition increases CDK4i/6i efficacy against therapy naïve and CDK4i/6i-resistant AM cells in xenograft and patient-derived xenograft (PDX) models and promotes a defective DNA repair, cell cycle arrested and apoptotic program. Notably, gene alterations poorly correlate with protein expression of cell cycle proteins in ALM or efficacy of CDK4i/6i, urging additional strategies when stratifying patients for CDK4i/6i trial inclusion. Concurrent targeting of the MAPK pathway and CDK4/6 represents a new approach for patients with metastatic ALM to improve outcomes., (© 2023. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
14. Immune landscape of tertiary lymphoid structures in hepatocellular carcinoma (HCC) treated with neoadjuvant immune checkpoint blockade.
- Author
-
Shu DH, Ho WJ, Kagohara LT, Girgis A, Shin SM, Danilova L, Lee JW, Sidiropoulos DN, Mitchell S, Munjal K, Howe K, Bendinelli KJ, Qi H, Mo G, Montagne J, Leatherman JM, Lopez-Vidal TY, Zhu Q, Huff AL, Yuan X, Hernandez A, Coyne EM, Zaidi N, Zabransky DJ, Engle LL, Ogurtsova A, Baretti M, Laheru D, Durham JN, Wang H, Anders R, Jaffee EM, Fertig EJ, and Yarchoan M
- Abstract
Neoadjuvant immunotherapy is thought to produce long-term remissions through induction of antitumor immune responses before removal of the primary tumor. Tertiary lymphoid structures (TLS), germinal center-like structures that can arise within tumors, may contribute to the establishment of immunological memory in this setting, but understanding of their role remains limited. Here, we investigated the contribution of TLS to antitumor immunity in hepatocellular carcinoma (HCC) treated with neoadjuvant immunotherapy. We found that neoadjuvant immunotherapy induced the formation of TLS, which were associated with superior pathologic response, improved relapse free survival, and expansion of the intratumoral T and B cell repertoire. While TLS in viable tumor displayed a highly active mature morphology, in areas of tumor regression we identified an involuted TLS morphology, which was characterized by dispersion of the B cell follicle and persistence of a T cell zone enriched for ongoing antigen presentation and T cell-mature dendritic cell interactions. Involuted TLS showed increased expression of T cell memory markers and expansion of CD8
+ cytotoxic and tissue resident memory clonotypes. Collectively, these data reveal the circumstances of TLS dissolution and suggest a functional role for late-stage TLS as sites of T cell memory formation after elimination of viable tumor., Competing Interests: Competing interests M.Y. reports grant/research support from Bristol-Myers Squibb, Incyte, Genentech (to Johns Hopkins) and honoraria from Genentech, Exelixis, Eisai, AstraZeneca, Replimune, Hepion, and equity in Adventris Pharmaceuticals. E.J.F is on the Scientific Advisory Board of Viosera/Reistance Bio, is a paid consultant for Merck and Mestag Therapeutics, and receives research funds from Abbvie. W.J.H. has received patent royalties from Rodeo/Amgen and is the recipient of grants from Sanofi, NeoTX, and CirclePharma. He has received speaking/travel honoraria from Exelixis and Standard BioTools. E.M.J. reports grant/research support from the Lustgarten Foundation, Break Through Cancer, Genentech, Bristol-Meyers Squibb; honoraria from Achilles, DragonFly, Parker Institute, Cancer Prevention and Research Institute of Texas, Surge, HDT Bio, Mestag Therapeutics, Medical Home Group; and equity in AbMeta Therapeutics and Adventris Pharmaceuticals. D.J.Z. reports grant/research support from Roche/Genentech.- Published
- 2023
- Full Text
- View/download PDF
15. Profiling of syngeneic mouse HCC tumor models as a framework to understand anti-PD-1 sensitive tumor microenvironments.
- Author
-
Zabransky DJ, Danilova L, Leatherman JM, Lopez-Vidal TY, Sanchez J, Charmsaz S, Gross NE, Shin S, Yuan X, Hernandez A, Yang H, Xavier S, Shu D, Saeed A, Munjal K, Kamdar Z, Kagohara LT, Jaffee EM, Yarchoan M, and Ho WJ
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Immunotherapy methods, Tumor Microenvironment, Programmed Cell Death 1 Receptor metabolism, Carcinoma, Hepatocellular pathology, Liver Neoplasms pathology
- Abstract
Background and Aims: The treatment of hepatocellular carcinoma (HCC) has been transformed by the use of immune checkpoint inhibitors. However, most patients with HCC do not benefit from treatment with immunotherapy. There is an urgent need to understand the mechanisms that underlie response or resistance to immunotherapy for patients with HCC. The use of syngeneic mouse models that closely recapitulate the heterogeneity of human HCC will provide opportunities to examine the complex interactions between cancer cells and nonmalignant cells in the tumor microenvironment., Approach and Results: We leverage a multifaceted approach that includes imaging mass cytometry and suspension cytometry by time of flight to profile the tumor microenvironments of the Hep53.4, Hepa 1-6, RIL-175, and TIBx (derivative of TIB-75) syngeneic mouse HCC models. The immune tumor microenvironments vary across these four models, and various immunosuppressive pathways exist at baseline in orthotopic liver tumors derived from these models. For instance, TIBx, which is resistant to anti-programmed cell death protein 1 therapy, contains a high proportion of "M2-like" tumor-associated macrophages with the potential to diminish antitumor immunity. Investigation of The Cancer Genome Atlas reveals that the baseline immunologic profiles of Hep53.4, RIL-175, and TIBx are broadly representative of human HCCs; however, Hepa 1-6 does not recapitulate the immune tumor microenvironment of the vast majority of human HCCs., Conclusions: There is a wide diversity in the immune tumor microenvironments in preclinical models and in human HCC, highlighting the need to use multiple syngeneic HCC models to improve the understanding of how to treat HCC through immune modulation., (Copyright © 2023 American Association for the Study of Liver Diseases.)
- Published
- 2023
- Full Text
- View/download PDF
16. Stromal changes in the aged lung induce an emergence from melanoma dormancy.
- Author
-
Fane ME, Chhabra Y, Alicea GM, Maranto DA, Douglass SM, Webster MR, Rebecca VW, Marino GE, Almeida F, Ecker BL, Zabransky DJ, Hüser L, Beer T, Tang HY, Kossenkov A, Herlyn M, Speicher DW, Xu W, Xu X, Jaffee EM, Aguirre-Ghiso JA, and Weeraratna AT
- Subjects
- Aged, Fibroblasts pathology, Humans, Neoplasm Invasiveness pathology, Neoplasm, Residual, Proto-Oncogene Proteins, Receptor Protein-Tyrosine Kinases, Skin pathology, Wnt-5a Protein, c-Mer Tyrosine Kinase, Axl Receptor Tyrosine Kinase, Aging pathology, Lung pathology, Melanoma pathology, Neoplasm Metastasis pathology, Stromal Cells pathology, Tumor Microenvironment
- Abstract
Disseminated cancer cells from primary tumours can seed in distal tissues, but may take several years to form overt metastases, a phenomenon that is termed tumour dormancy. Despite its importance in metastasis and residual disease, few studies have been able to successfully characterize dormancy within melanoma. Here we show that the aged lung microenvironment facilitates a permissive niche for efficient outgrowth of dormant disseminated cancer cells-in contrast to the aged skin, in which age-related changes suppress melanoma growth but drive dissemination. These microenvironmental complexities can be explained by the phenotype switching model, which argues that melanoma cells switch between a proliferative cell state and a slower-cycling, invasive state
1-3 . It was previously shown that dermal fibroblasts promote phenotype switching in melanoma during ageing4-8 . We now identify WNT5A as an activator of dormancy in melanoma disseminated cancer cells within the lung, which initially enables the efficient dissemination and seeding of melanoma cells in metastatic niches. Age-induced reprogramming of lung fibroblasts increases their secretion of the soluble WNT antagonist sFRP1, which inhibits WNT5A in melanoma cells and thereby enables efficient metastatic outgrowth. We also identify the tyrosine kinase receptors AXL and MER as promoting a dormancy-to-reactivation axis within melanoma cells. Overall, we find that age-induced changes in distal metastatic microenvironments promote the efficient reactivation of dormant melanoma cells in the lung., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
17. Clinical, Genomic, and Transcriptomic Data Profiling of Biliary Tract Cancer Reveals Subtype-Specific Immune Signatures.
- Author
-
Mody K, Jain P, El-Refai SM, Azad NS, Zabransky DJ, Baretti M, Shroff RT, Kelley RK, El-Khouiery AB, Hockenberry AJ, Lau D, Lesinski GB, and Yarchoan M
- Subjects
- Bile Ducts, Intrahepatic pathology, Genomics, Humans, Proto-Oncogene Proteins p21(ras) genetics, Transcriptome genetics, Tumor Microenvironment genetics, Bile Duct Neoplasms genetics, Biliary Tract Neoplasms genetics, Cholangiocarcinoma genetics, Gallbladder Neoplasms genetics
- Abstract
Purpose: Biliary tract cancers (BTCs) are aggressive cancers that carry a poor prognosis. An enhanced understanding of the immune landscape of anatomically and molecularly defined subsets of BTC may improve patient selection for immunotherapy and inform immune-based combination treatment strategies., Methods: We analyzed deidentified clinical, genomic, and transcriptomic data from the Tempus database to determine the mutational frequency and mutational clustering across the three major BTC subtypes (intrahepatic cholangiocarcinoma [IHC], extrahepatic cholangiocarcinoma, and gallbladder cancer). We subsequently determined the relationship between specific molecular alterations and anatomical subsets and features of the BTC immune microenvironment., Results: We analyzed 454 samples of BTC, of which the most commonly detected alterations were TP53 (42.5%), CDKN2A (23.4%), ARID1A (19.6%), BAP1 (15.5%), KRAS (15%), CDKN2B (14.2%), PBRM1 (11.7%), IDH1 (11.7%), TERT (8.4%), KMT2C (10.4%) and LRP1B (8.4%), and FGFR2 fusions (8.7%). Potentially actionable molecular alterations were identified in 30.5% of BTCs including 39.1% of IHC. Integrative cluster analysis revealed four distinct molecular clusters, with cluster 4 predominately associated with FGFR2 rearrangements and BAP1 mutations in IHC. Immune-related biomarkers indicative of an inflamed tumor-immune microenvironment were elevated in gallbladder cancers and in cluster 1, which was enriched for TP53 , KRAS , and ATM mutations. Multiple common driver genes, including TP53 , FGFR2 , IDH1 , TERT , BRAF , and BAP1 , were individually associated with unique BTC immune microenvironments., Conclusion: BTC subtypes exhibit diverse DNA alterations, RNA inflammatory signatures, and immune biomarkers. The association between specific BTC anatomical subsets, molecular alterations, and immunophenotypes highlights new opportunities for therapeutic development., Competing Interests: Kabir ModyStock and Other Ownership Interests: CytoDyn, ONCOtherapeuticsConsulting or Advisory Role: Celgene, Genentech/Roche, Merrimack, Eisai, AstraZeneca, Vicus Therapeutics, Ipsen, Boston Scientific, BTG, BTG, Exelixis, Incyte (Inst), QED TherapeuticsResearch Funding: FibroGen (Inst), Senhwa Biosciences (Inst), ARIAD (Inst), TRACON Pharma (Inst), MedImmune (Inst), Agios (Inst), ArQule (Inst), Taiho Pharmaceutical (Inst), Gritstone Bio (Inst), Incyte (Inst), Merck (Inst), Vyriad (Inst), Turnstone Bio (Inst), AstraZeneca (Inst), Basilea (Inst) Prerna JainEmployment: Tempus, Concerto HealthAITravel, Accommodations, Expenses: Tempus Sherif M. El-RefaiEmployment: TempusStock and Other Ownership Interests: Tempus Nilofer S. AzadConsulting or Advisory Role: QED Therapeutics, Merck, Incyte, Helsinn Therapeutics/QED Therapeutics, AstraZeneca, Mirati TherapeuticsResearch Funding: Celgene (Inst), Genentech (Inst), Astex Pharmaceuticals (Inst), Agios (Inst), Merck (Inst), Bristol Myers Squibb (Inst), Syndax (Inst), Array BioPharma (Inst), Intensity Therapeutics (Inst), Bayer (Inst), EMD Serono, Debiopharm Group (Inst), Incyte (Inst), Loxo/Lilly (Inst), AtlasMedx (Inst) Daniel J. ZabranskyHonoraria: Remedy Health Group, LLCResearch Funding: Roche/GenentechPatents, Royalties, Other Intellectual Property: Under separate licensing agreements between Horizon Discovery, Ltd and The Johns Hopkins University, I am entitled to a share of royalties received by the university on sales of products Rachna T. ShroffConsulting or Advisory Role: Exelixis, Merck, QED Therapeutics, Incyte, AstraZeneca, Taiho Pharmaceutical, Boehringer Ingelheim, Servier, Genentech, BasileaResearch Funding: Pieris Pharmaceuticals, Taiho Pharmaceutical, Merck, Exelixis, QED Therapeutics, Rafael Pharmaceuticals, Bristol Myers Squibb, Bayer, Immunovaccine, Seattle Genetics, Novocure, NuCana, Loxo/Lilly, Faeth Therapeutics R. Katie KelleyConsulting or Advisory Role: Agios (Inst), AstraZeneca (Inst), Bristol Myers Squibb (Inst), Genentech/Roche, Merck (Inst), Gilead Sciences, Exact Sciences, Kinnate Biopharma, Exelixis/Ipsen (Inst)Research Funding: Lilly (Inst), Exelixis (Inst), Novartis (Inst), Bristol Myers Squibb (Inst), MedImmune (Inst), Merck Sharp & Dohme (Inst), Agios (Inst), AstraZeneca (Inst), Adaptimmune (Inst), Taiho Pharmaceutical (Inst), Bayer (Inst), QED Therapeutics (Inst), EMD Serono (Inst), Partner Therapeutics (Inst), Genentech/Roche (Inst), Surface Oncology (Inst), Relay Therapeutics (Inst), Loxo/Lilly (Inst)Travel, Accommodations, Expenses: Ipsen Anthony B. El-KhouieryHonoraria: Bayer, Bristol Myers Squibb, Roche/Genentech, EMD Serono, Eisai, Merck, Agenus, Pieris Pharmaceuticals, Exelixis, CytomX Therapeutics, Gilead Sciences, AstraZeneca/MedImmune, ABL Bio, QED Therapeutics, Servier, Tallac TherapeuticsConsulting or Advisory Role: CytomX Therapeutics, Bristol Myers Squibb, Bayer, Eisai, Roche, EMD Serono, Merck, Exelixis, Pieris Pharmaceuticals, Agenus, Gilead Sciences, AstraZeneca/MedImmune, ABL Bio, QED Therapeutics, Servier, Tallac TherapeuticsResearch Funding: AstraZeneca, Astex Pharmaceuticals, Fulgent Genetics Adam J. HockenberryEmployment: TempusStock and Other Ownership Interests: Tempus Denise LauEmployment: TempusStock and Other Ownership Interests: TempusPatents, Royalties, Other Intellectual Property: Tempus Gregory B. LesinskiConsulting or Advisory Role: ProDaResearch Funding: Merck (Inst), Bristol Myers Squibb (Inst), Novartis (Inst), Boehringer Ingelheim (Inst), Vaccinex (Inst) Mark YarchoanConsulting or Advisory Role: Eisai, Exelixis, AstraZeneca, Genentech/Roche, Replimune, Hepion PharmaceuticalsResearch Funding: Bristol Myers Squibb (Inst), Merck (Inst), Exelixis (Inst), Incyte (Inst)Uncompensated Relationships: MerckNo other potential conflicts of interest were reported.
- Published
- 2022
- Full Text
- View/download PDF
18. Shared genetic and epigenetic changes link aging and cancer.
- Author
-
Zabransky DJ, Jaffee EM, and Weeraratna AT
- Subjects
- Aged, Aging genetics, Aging pathology, Epigenesis, Genetic genetics, Epigenome, Humans, Tumor Microenvironment genetics, DNA Methylation, Neoplasms genetics, Neoplasms therapy
- Abstract
Aging is a universal biological process that increases the risk of multiple diseases including cancer. Growing evidence shows that alterations in the genome and epigenome, driven by similar mechanisms, are found in both aged cells and cancer cells. In this review, we detail the genetic and epigenetic changes associated with normal aging and the mechanisms responsible for these changes. By highlighting genetic and epigenetic alterations in the context of tumorigenesis, cancer progression, and the aging tumor microenvironment, we examine the possible impacts of the normal aging process on malignant transformation. Finally, we examine the implications of age-related genetic and epigenetic alterations in both tumors and patients for the treatment of cancer., Competing Interests: Declaration of interests D.J.Z. declares no competing interests. E.M.J. is a paid consultant for Adaptive Biotechnologies, CSTONE, Achilles, DragonFly, Candel Therapeutics, NextCure Biotech, and Genocea. She receives funding from Lustgarten Foundation and Bristol Myers Squibb. She is the Chief Medical Advisor for Lustgarten and SAB advisor to the Parker Institute for Cancer Immunotherapy (PICI) and for the C3 Cancer Institute. She is a founding member of Abmeta. A.T.W. is a member of the board of directors of ReGAIN Therapeutics and is on the scientific advisory board of PhoreMost Technologies., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
19. NOTCH1 PEST domain variants are responsive to standard of care treatments despite distinct transformative properties in a breast cancer model.
- Author
-
Cravero K, Pantone MV, Shin DH, Bergman R, Cochran R, Chu D, Zabransky DJ, Karthikeyan S, Waters IG, Hunter N, Rosen DM, Kyker-Snowman K, Dalton WB, Button B, Shinn D, Wong HY, Donaldson J, Hurley PJ, Croessmann S, and Park BH
- Subjects
- Cell Line, Tumor, Cell Proliferation genetics, Gamma Secretase Inhibitors and Modulators, Humans, Receptor, Notch1 genetics, Receptor, Notch1 metabolism, Signal Transduction, Standard of Care, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms therapy
- Abstract
Activating variants in the PEST region of NOTCH1 have been associated with aggressive phenotypes in human cancers, including triple-negative breast cancer (TNBC). Previous studies suggested that PEST domain variants in TNBC patients resulted in increased cell proliferation, invasiveness, and decreased overall survival. In this study, we assess the phenotypic transformation of activating NOTCH1 variants and their response to standard of care therapies. AAV-mediated gene targeting was used to isogenically incorporate 3 NOTCH1 variants, including a novel TNBC frameshift variant, in two non-tumorigenic breast epithelial cell lines, MCF10A and hTERT-IMEC. Two different variants at the NOTCH1 A2241 site (A2441fs and A2441T) both demonstrated increased transformative properties when compared to a non-transformative PEST domain variant (S2523L). These phenotypic changes include proliferation, migration, anchorage-independent growth, and MAPK pathway activation. In contrast to previous studies, activating NOTCH1 variants did not display sensitivity to a gamma secretase inhibitor (GSI) or resistance to chemotherapies. This study demonstrates distinct transformative phenotypes are specific to a given variant within NOTCH1 and these phenotypes do not correlate with sensitivities or resistance to chemotherapies or GSIs. Although previous studies have suggested NOTCH1 variants may be prognostic for TNBC, our study does not demonstrate prognostic ability of these variants and suggests further characterization would be required for clinical applications., Competing Interests: CONFLICTS OF INTEREST B.H.P. is a paid consultant for Jackson Labs, EQRx, Sermonix, Hologics, Guardant Health and is a paid scientific advisory board member for Celcuity Inc. B.H.P. also has research contracts with GE Healthcare, Lilly and Pfizer. Under separate licensing agreements between Horizon Discovery, LTD and The Johns Hopkins University, B.H.P. is entitled to a share of royalties received by the University on sales of products. The terms of this arrangement are being managed by the Johns Hopkins University in accordance with its conflict of interest policies. J.D. receives royalties from patents held by City of Hope and Thomas Jefferson University. J.D. also holds stock awards from Xilio Therapeutics. None of these listed had do direct conflict of interest on the project., (Copyright: © 2022 Cravero et al.)
- Published
- 2022
- Full Text
- View/download PDF
20. Implantation of a neoantigen-targeted hydrogel vaccine prevents recurrence of pancreatic adenocarcinoma after incomplete resection.
- Author
-
Delitto D, Zabransky DJ, Chen F, Thompson ED, Zimmerman JW, Armstrong TD, Leatherman JM, Suri R, Lopez-Vidal TY, Huff AL, Lyman MR, Guinn SR, Baretti M, Kagohara LT, Ho WJ, Azad NS, Burns WR, He J, Wolfgang CL, Burkhart RA, Zheng L, Yarchoan M, Zaidi N, and Jaffee EM
- Subjects
- Animals, Hydrogels, Immunotherapy, Mice, Tumor Microenvironment, Adenocarcinoma prevention & control, Adenocarcinoma surgery, Cancer Vaccines, Pancreatic Neoplasms
- Abstract
Tumor involvement of major vascular structures limits surgical options in pancreatic adenocarcinoma (PDAC), which in turn limits opportunities for cure. Despite advances in locoregional approaches, there is currently no role for incomplete resection. This study evaluated a gelatinized neoantigen-targeted vaccine applied to a grossly positive resection margin in preventing local recurrence. Incomplete surgical resection was performed in mice bearing syngeneic flank Panc02 tumors, leaving a 1 mm rim adherent to the muscle bed. A previously validated vaccine consisting of neoantigen peptides, a stimulator of interferon genes (STING) agonist and AddaVax
TM (termed PancVax) was embedded in a hyaluronic acid hydrogel and applied to the tumor bed. Tumor remnants, regional lymph nodes, and spleens were analyzed using histology, flow cytometry, gene expression profiling, and ELISPOT assays. The immune microenvironment at the tumor margin after surgery alone was characterized by a transient influx of myeloid-derived suppressor cells (MDSCs), prolonged neutrophil influx, and near complete loss of cytotoxic T cells. Application of PancVax gel was associated with enhanced T cell activation in the draining lymph node and expansion of neoantigen-specific T cells in the spleen. Mice implanted with PancVax gel demonstrated no evidence of residual tumor at two weeks postoperatively and healed incisions at two months postoperatively without local recurrence. In summary, application of PancVax gel at a grossly positive tumor margin led to systemic expansion of neoantigen-specific T cells and effectively prevented local recurrence. These findings support further work into locoregional adjuncts to immune modulation in PDAC., Competing Interests: WH is a co-inventor of patents with potential for receiving royalties from Rodeo Therapeutics. MY received research grants from Incyte, Bristol-Myers Squibb, Exelixis, and is a consultant for AstraZeneca, Eisai, Exelixis, and Genentech. EJ received a commercial research grant from Bristol-Myers Squibb and Aduro Biotech, and is a consultant and advisory board member for Lustgarten Foundation, Parker Institute for Cancer Immunotherapy, CStone, Dragonfly, Genocea, Achilles and Adaptive Biotechnologies, and co-founder of Abmeta Biotech., (© 2021 The Author(s). Published with license by Taylor & Francis Group, LLC.)- Published
- 2021
- Full Text
- View/download PDF
21. Hierarchical tumor heterogeneity mediated by cell contact between distinct genetic subclones.
- Author
-
Karthikeyan S, Waters IG, Dennison L, Chu D, Donaldson J, Shin DH, Rosen DM, Gonzalez-Ericsson PI, Sanchez V, Sanders ME, Pantone MV, Bergman RE, Davidson BA, Reed SC, Zabransky DJ, Cravero K, Kyker-Snowman K, Button B, Wong HY, Hurley PJ, Croessmann S, and Park BH
- Subjects
- Breast Neoplasms metabolism, Cell Communication genetics, Cell Line, Tumor, Cell Transformation, Neoplastic genetics, Class I Phosphatidylinositol 3-Kinases genetics, Coculture Techniques, Female, Fibronectins antagonists & inhibitors, Fibronectins genetics, Fibronectins metabolism, Gene Expression Regulation, Neoplastic, Gene Frequency, Gene Knockout Techniques, Humans, Immunohistochemistry, MCF-7 Cells, Mutation, Phenotype, Receptor, ErbB-2 genetics, Breast Neoplasms genetics, Breast Neoplasms pathology
- Abstract
Intratumor heterogeneity is an important mediator of poor outcomes in many cancers, including breast cancer. Genetic subclones frequently contribute to this heterogeneity; however, their growth dynamics and interactions remain poorly understood. PIK3CA and HER2 alterations are known to coexist in breast and other cancers. Herein, we present data that describe the ability of oncogenic PIK3CA mutant cells to induce the proliferation of quiescent HER2 mutant cells through a cell contact-mediated mechanism. Interestingly, the HER2 cells proliferated to become the major subclone over PIK3CA counterparts both in vitro and in vivo. Furthermore, this phenotype was observed in both hormone receptor-positive and -negative cell lines, and was dependent on the expression of fibronectin from mutant PIK3CA cells. Analysis of human tumors demonstrated similar HER2:PIK3CA clonal dynamics and fibronectin expression. Our study provides insight into nonrandom subclonal architecture of heterogenous tumors, which may aid the understanding of tumor evolution and inform future strategies for personalized medicine.
- Published
- 2021
- Full Text
- View/download PDF
22. Hotspot SF3B1 mutations induce metabolic reprogramming and vulnerability to serine deprivation.
- Author
-
Dalton WB, Helmenstine E, Walsh N, Gondek LP, Kelkar DS, Read A, Natrajan R, Christenson ES, Roman B, Das S, Zhao L, Leone RD, Shinn D, Groginski T, Madugundu AK, Patil A, Zabransky DJ, Medford A, Lee J, Cole AJ, Rosen M, Thakar M, Ambinder A, Donaldson J, DeZern AE, Cravero K, Chu D, Madero-Marroquin R, Pandey A, Hurley PJ, Lauring J, and Park BH
- Subjects
- Animals, Cell Line, Tumor, Energy Metabolism genetics, Glycine, Humans, Mice, Neoplasm Proteins genetics, Phosphoglycerate Dehydrogenase genetics, Phosphoglycerate Dehydrogenase metabolism, Proteome genetics, Xenograft Model Antitumor Assays, Cellular Reprogramming, Mutation, Neoplasm Proteins metabolism, Neoplasms diet therapy, Neoplasms genetics, Neoplasms metabolism, Neoplasms pathology, Phosphoproteins genetics, Phosphoproteins metabolism, Proteome metabolism, RNA Splicing Factors genetics, RNA Splicing Factors metabolism, Serine, Transcriptome
- Abstract
Cancer-associated mutations in the spliceosome gene SF3B1 create a neomorphic protein that produces aberrant mRNA splicing in hundreds of genes, but the ensuing biologic and therapeutic consequences of this missplicing are not well understood. Here we have provided evidence that aberrant splicing by mutant SF3B1 altered the transcriptome, proteome, and metabolome of human cells, leading to missplicing-associated downregulation of metabolic genes, decreased mitochondrial respiration, and suppression of the serine synthesis pathway. We also found that mutant SF3B1 induces vulnerability to deprivation of the nonessential amino acid serine, which was mediated by missplicing-associated downregulation of the serine synthesis pathway enzyme PHGDH. This vulnerability was manifest both in vitro and in vivo, as dietary restriction of serine and glycine in mice was able to inhibit the growth of SF3B1MUT xenografts. These findings describe a role for SF3B1 mutations in altered energy metabolism, and they offer a new therapeutic strategy against SF3B1MUT cancers.
- Published
- 2019
- Full Text
- View/download PDF
23. The estrogen receptor-alpha S118P variant does not affect breast cancer incidence or response to endocrine therapies.
- Author
-
Button B, Croessmann S, Chu D, Rosen DM, Zabransky DJ, Dalton WB, Cravero K, Kyker-Snowman K, Waters I, Karthikeyan S, Christenson ES, Donaldson J, Hunter T, Dennison L, Ramin C, May B, Roden R, Petry D, Armstrong DK, Visvanathan K, and Park BH
- Subjects
- Adult, Aged, Breast Neoplasms genetics, Case-Control Studies, Cell Line, Tumor, Cell Movement, Cell Proliferation, Estrogen Receptor alpha metabolism, Female, Fulvestrant therapeutic use, Genetic Variation, Humans, Incidence, MCF-7 Cells, Middle Aged, Phosphorylation, Survival Analysis, Tamoxifen therapeutic use, Treatment Outcome, Antineoplastic Agents, Hormonal therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms epidemiology, Estrogen Receptor alpha genetics, Germ-Line Mutation
- Abstract
Purpose: Estrogen receptor-alpha (ER) is a therapeutic target of ER-positive (ER+) breast cancers. Although ER signaling is complex, many mediators of this pathway have been identified. Specifically, phosphorylation of ER at serine 118 affects responses to estrogen and therapeutic ligands and has been correlated with clinical outcomes in ER+ breast cancer patients. We hypothesized that a newly described germline variant (S118P) at this residue would drive cellular changes consistent with breast cancer development and/or hormone resistance., Methods: Isogenic human breast epithelial cell line models harboring ER S118P were developed via genome editing and characterized to determine the functional effects of this variant. We also examined the frequency of ER S118P in a case-control study (N = 536) of women with and without breast cancer with a familial risk., Results: In heterozygous knock-in models, the S118P variant demonstrated no significant change in proliferation, migration, MAP Kinase pathway signaling, or response to the endocrine therapies tamoxifen and fulvestrant. Further, there was no difference in the prevalence of S118P between women with and without cancer relative to population registry databases., Conclusions: This study suggests that the ER S118P variant does not affect risk for breast cancer or hormone therapy resistance. Germline screening and modification of treatments for patients harboring this variant are likely not warranted.
- Published
- 2019
- Full Text
- View/download PDF
24. HER2 Reactivation through Acquisition of the HER2 L755S Mutation as a Mechanism of Acquired Resistance to HER2-targeted Therapy in HER2 + Breast Cancer.
- Author
-
Xu X, De Angelis C, Burke KA, Nardone A, Hu H, Qin L, Veeraraghavan J, Sethunath V, Heiser LM, Wang N, Ng CKY, Chen ES, Renwick A, Wang T, Nanda S, Shea M, Mitchell T, Rajendran M, Waters I, Zabransky DJ, Scott KL, Gutierrez C, Nagi C, Geyer FC, Chamness GC, Park BH, Shaw CA, Hilsenbeck SG, Rimawi MF, Gray JW, Weigelt B, Reis-Filho JS, Osborne CK, and Schiff R
- Subjects
- Afatinib, Animals, Antibodies, Monoclonal, Humanized administration & dosage, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Line, Tumor, Cell Proliferation drug effects, Female, Humans, Lapatinib, Mice, Mutation, Quinazolines administration & dosage, Quinazolines adverse effects, Receptor, ErbB-2 antagonists & inhibitors, Receptors, Estrogen genetics, Signal Transduction drug effects, Trastuzumab administration & dosage, Trastuzumab adverse effects, Xenograft Model Antitumor Assays, Breast Neoplasms drug therapy, Drug Resistance, Neoplasm genetics, Molecular Targeted Therapy, Receptor, ErbB-2 genetics
- Abstract
Purpose: Resistance to anti-HER2 therapies in HER2
+ breast cancer can occur through activation of alternative survival pathways or reactivation of the HER signaling network. Here we employed BT474 parental and treatment-resistant cell line models to investigate a mechanism by which HER2+ breast cancer can reactivate the HER network under potent HER2-targeted therapies. Experimental Design: Resistant derivatives to lapatinib (L), trastuzumab (T), or the combination (LR/TR/LTR) were developed independently from two independent estrogen receptor ER+ /HER2+ BT474 cell lines (AZ/ATCC). Two derivatives resistant to the lapatinib-containing regimens (BT474/AZ-LR and BT474/ATCC-LTR lines) that showed HER2 reactivation at the time of resistance were subjected to massive parallel sequencing and compared with parental lines. Ectopic expression and mutant-specific siRNA interference were applied to analyze the mutation functionally. In vitro and in vivo experiments were performed to test alternative therapies for mutant HER2 inhibition. Results: Genomic analyses revealed that the HER2 L755S mutation was the only common somatic mutation gained in the BT474/AZ-LR and BT474/ATCC-LTR lines. Ectopic expression of HER2 L755S induced acquired lapatinib resistance in the BT474/AZ, SK-BR-3, and AU565 parental cell lines. HER2 L755S-specific siRNA knockdown reversed the resistance in BT474/AZ-LR and BT474/ATCC-LTR lines. The HER1/2-irreversible inhibitors afatinib and neratinib substantially inhibited both resistant cell growth and the HER2 and downstream AKT/MAPK signaling driven by HER2 L755S in vitro and in vivo Conclusions: HER2 reactivation through acquisition of the HER2 L755S mutation was identified as a mechanism of acquired resistance to lapatinib-containing HER2-targeted therapy in preclinical HER2-amplified breast cancer models, which can be overcome by irreversible HER1/2 inhibitors. Clin Cancer Res; 23(17); 5123-34. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
25. Whole-Exome Sequencing of Metaplastic Breast Carcinoma Indicates Monoclonality with Associated Ductal Carcinoma Component.
- Author
-
Avigdor BE, Beierl K, Gocke CD, Zabransky DJ, Cravero K, Kyker-Snowman K, Button B, Chu D, Croessmann S, Cochran RL, Connolly RM, Park BH, Wheelan SJ, and Cimino-Mathews A
- Subjects
- Adult, Aged, Aged, 80 and over, Breast pathology, Breast Neoplasms metabolism, Breast Neoplasms pathology, Carcinoma, Ductal, Breast metabolism, Carcinoma, Ductal, Breast pathology, Clone Cells metabolism, Clone Cells pathology, DNA Copy Number Variations, Female, Humans, Metaplasia genetics, Metaplasia metabolism, Middle Aged, Mutation, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms metabolism, Triple Negative Breast Neoplasms pathology, Breast metabolism, Breast Neoplasms genetics, Carcinoma, Ductal, Breast genetics, Exome Sequencing methods
- Abstract
Purpose: Although most human cancers display a single histology, there are unusual cases where two or more distinct tissue types present within a primary tumor. One such example is metaplastic breast carcinoma, a rare but aggressive cancer with a heterogeneous histology, including squamous, chondroid, and spindle cells. Metaplastic carcinomas often contain an admixed conventional ductal invasive or in situ mammary carcinoma component, and are typically triple-negative for estrogen receptor, progesterone receptor, and HER-2 amplification/overexpression. An unanswered question is the origin of metaplastic breast cancers. While they may arise independently from their ductal components, their close juxtaposition favors a model that postulates a shared origin, either as two derivatives from the same primary cancer or one histology as an outgrowth of the other. Understanding the mechanism of development of these tumors may inform clinical decisions. Experimental Design: We performed exome sequencing for paired metaplastic and adjacent conventional invasive ductal carcinomas in 8 patients and created a pipeline to identify somatic variants and predict their functional impact, without having normal tissue. We then determined the genetic relationships between the histologically distinct compartments. Results: In each case, the tumor components have nearly identical landscapes of somatic mutation, implying that the differing histologies do not derive from genetic clonal divergence. Conclusions: A shared origin for tumors with differing histologies suggests that epigenetic or noncoding changes may mediate the metaplastic phenotype and that alternative therapeutic approaches, including epigenetic therapies, may be required for metaplastic breast cancers. Clin Cancer Res; 23(16); 4875-84. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
26. Single-Nucleotide Polymorphism Leading to False Allelic Fraction by Droplet Digital PCR.
- Author
-
Christenson ES, Dalton WB, Chu D, Waters I, Cravero K, Zabransky DJ, DeZern AE, and Park BH
- Subjects
- DNA, Neoplasm blood, False Positive Reactions, Humans, Mutation, Alleles, DNA, Neoplasm genetics, Phosphoproteins genetics, Polymerase Chain Reaction methods, Polymorphism, Single Nucleotide genetics, RNA Splicing Factors genetics
- Abstract
Background: Molecular-based diagnostics have great utility for cancer detection. We have used droplet digital PCR (ddPCR) as a platform for identifying mutations in circulating plasma tumor DNA (ptDNA). We present the unexpected finding of a spurious mutant allele fraction that was discovered to be artifactual because of the presence of a single-nucleotide polymorphism (SNP) in a patient sample., Design and Methods: Probe and primer combinations for the K700 and V701 loci of the SF3B1 spliceosome gene were designed for ddPCR to identify the percentage of mutant and wild-type alleles. Clinical samples from patients with cancer with known SF3B1 mutations were collected and tested to evaluate the assays' ability to detect SF3B1 mutations in ptDNA., Results: Patient samples showed SF3B1 K700E mutations within the ptDNA of 4 patients with acute leukemia and 3 with myelodysplastic syndrome who were known to harbor this mutation. A blood sample from a patient with lung cancer with a known SF3B1 V701F mutation was also analyzed and this mutation was successfully identified in ptDNA. However, 1 of the patients with a K700E mutation was found to have a mutational burden of 98%. After careful analysis of this locus by Sanger sequencing and ddPCR, this patient was found to have an SNP (R702R), which prevented binding of the ddPCR wild-type probe to its cognate allele., Conclusions: These results further support that ddPCR-based assays may be valuable companion diagnostics for the identification and monitoring of patients with cancer, but the results also emphasize the need to identify SNPs at loci that are being analyzed., (© 2017 American Association for Clinical Chemistry.)
- Published
- 2017
- Full Text
- View/download PDF
27. PIK3CA mutations and TP53 alterations cooperate to increase cancerous phenotypes and tumor heterogeneity.
- Author
-
Croessmann S, Wong HY, Zabransky DJ, Chu D, Rosen DM, Cidado J, Cochran RL, Dalton WB, Erlanger B, Cravero K, Button B, Kyker-Snowman K, Hurley PJ, Lauring J, and Park BH
- Subjects
- Animals, Cell Cycle, Cell Line, Tumor, Cell Proliferation, Centromere genetics, DNA Copy Number Variations, Disease Models, Animal, Drug Resistance, Neoplasm genetics, Female, Gene Amplification, Gene Editing, Gene Knockout Techniques, Genomic Instability, Genotype, Humans, Mice, Paclitaxel pharmacology, Breast Neoplasms genetics, Breast Neoplasms pathology, Class I Phosphatidylinositol 3-Kinases genetics, Mutation, Phenotype, Tumor Suppressor Protein p53 genetics
- Abstract
Background/purpose: The combined contributions of oncogenes and tumor suppressor genes toward carcinogenesis remain poorly understood. Elucidation of cancer gene cooperativity can provide new insights leading to more effective use of therapies., Experimental Design/methods: We used somatic cell genome editing to introduce singly and in combination PIK3CA mutations (E545K or H1047R) with TP53 alterations (R248W or knockout), to assess any enhanced cancerous phenotypes. The non-tumorigenic human breast epithelial cell line, MCF10A, was used as the parental cell line, and resultant cells were assessed via various in vitro assays, growth as xenografts, and drug sensitivity assays using targeted agents and chemotherapies., Results: Compared to single-gene-targeted cells and parental controls, cells with both a PIK3CA mutation and TP53 alteration had increased cancerous phenotypes including cell proliferation, soft agar colony formation, aberrant morphology in acinar formation assays, and genomic heterogeneity. Cells also displayed varying sensitivities to anti-neoplastic drugs, although all cells with PIK3CA mutations showed a relative increased sensitivity to paclitaxel. All cell lines remained non-tumorigenic., Conclusions: This cell line panel provides a resource for further elucidating cooperative genetic mediators of carcinogenesis and response to therapies.
- Published
- 2017
- Full Text
- View/download PDF
28. Individualized Molecular Analyses Guide Efforts (IMAGE): A Prospective Study of Molecular Profiling of Tissue and Blood in Metastatic Triple-Negative Breast Cancer.
- Author
-
Parsons HA, Beaver JA, Cimino-Mathews A, Ali SM, Axilbund J, Chu D, Connolly RM, Cochran RL, Croessmann S, Clark TA, Gocke CD, Jeter SC, Kennedy MR, Lauring J, Lee J, Lipson D, Miller VA, Otto GA, Rosner GL, Ross JS, Slater S, Stephens PJ, VanDenBerg DA, Wolff AC, Young LE, Zabransky DJ, Zhang Z, Zorzi J, Stearns V, and Park BH
- Subjects
- Adult, Aged, Biopsy, Drug Therapy, Female, Gene Expression Regulation, Neoplastic drug effects, High-Throughput Nucleotide Sequencing, Humans, Middle Aged, Mutation, Neoplasm Metastasis, Neoplasm Proteins biosynthesis, Precision Medicine, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms pathology, Biomarkers, Tumor blood, DNA, Neoplasm blood, Neoplasm Proteins genetics, Triple Negative Breast Neoplasms blood
- Abstract
Purpose: The clinical utility of next-generation sequencing (NGS) in breast cancer has not been demonstrated. We hypothesized that we could perform NGS of a new biopsy from patients with metastatic triple-negative breast cancer (TNBC) in a clinically actionable timeframe., Experimental Design: We planned to enroll 40 patients onto a prospective study, Individualized Molecular Analyses Guide Efforts (IMAGE), to evaluate the feasibility of obtaining a new biopsy of a metastatic site, perform NGS (FoundationOne), and convene a molecular tumor board to formulate treatment recommendations within 28 days. We collected blood at baseline and at time of restaging to assess cell-free circulating plasma tumor DNA (ptDNA)., Results: We enrolled 26 women with metastatic TNBC who had received ≥1 line of prior chemotherapy, and 20 (77%) underwent NGS of a metastatic site biopsy. Twelve (60%) evaluable patients received treatment recommendations within 28 days of consent. The study closed after 20 patients underwent NGS, based on protocol-specified interim futility analysis. Three patients went on to receive genomically directed therapies. Twenty-four of 26 patients had genetic alterations successfully detected in ptDNA. Among 5 patients, 4 mutations found in tumor tissues were not identified in blood, and 4 mutations found in blood were not found in corresponding tumors. In 9 patients, NGS of follow-up blood samples showed 100% concordance with baseline blood samples., Conclusions: This study demonstrates challenges of performing NGS on prospective tissue biopsies in patients with metastatic TNBC within 28 days, while also highlighting the potential use of blood as a more time-efficient and less invasive method of mutational assessment. Clin Cancer Res; 23(2); 379-86. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
29. ESR1 Mutations in Circulating Plasma Tumor DNA from Metastatic Breast Cancer Patients.
- Author
-
Chu D, Paoletti C, Gersch C, VanDenBerg DA, Zabransky DJ, Cochran RL, Wong HY, Toro PV, Cidado J, Croessmann S, Erlanger B, Cravero K, Kyker-Snowman K, Button B, Parsons HA, Dalton WB, Gillani R, Medford A, Aung K, Tokudome N, Chinnaiyan AM, Schott A, Robinson D, Jacks KS, Lauring J, Hurley PJ, Hayes DF, Rae JM, and Park BH
- Subjects
- Adult, Aged, Breast Neoplasms blood, Breast Neoplasms pathology, DNA Mutational Analysis, DNA, Neoplasm genetics, Female, Gene Frequency, Humans, Liver Neoplasms blood, Liver Neoplasms secondary, Middle Aged, Mutation, Missense, Breast Neoplasms genetics, DNA, Neoplasm blood, Estrogen Receptor alpha genetics, Liver Neoplasms genetics
- Abstract
Purpose: Mutations in the estrogen receptor (ER)α gene, ESR1, have been identified in breast cancer metastases after progression on endocrine therapies. Because of limitations of metastatic biopsies, the reported frequency of ESR1 mutations may be underestimated. Here, we show a high frequency of ESR1 mutations using circulating plasma tumor DNA (ptDNA) from patients with metastatic breast cancer., Experimental Design: We retrospectively obtained plasma samples from eight patients with known ESR1 mutations and three patients with wild-type ESR1 identified by next-generation sequencing (NGS) of biopsied metastatic tissues. Three common ESR1 mutations were queried for using droplet digital PCR (ddPCR). In a prospective cohort, metastatic tissue and plasma were collected contemporaneously from eight ER-positive and four ER-negative patients. Tissue biopsies were sequenced by NGS, and ptDNA ESR1 mutations were analyzed by ddPCR., Results: In the retrospective cohort, all corresponding mutations were detected in ptDNA, with two patients harboring additional ESR1 mutations not present in their metastatic tissues. In the prospective cohort, three ER-positive patients did not have adequate tissue for NGS, and no ESR1 mutations were identified in tissue biopsies from the other nine patients. In contrast, ddPCR detected seven ptDNA ESR1 mutations in 6 of 12 patients (50%)., Conclusions: We show that ESR1 mutations can occur at a high frequency and suggest that blood can be used to identify additional mutations not found by sequencing of a single metastatic lesion., Competing Interests: of Potential Conflicts of Interest: B.H.P. is a paid consultant for Novartis and is a member of the scientific advisory boards of Horizon Discovery, LTD and Loxo Oncology, and has research contracts with Genomic Health, Inc and Foundation Medicine. Under separate licensing agreements between Horizon Discovery, LTD and The Johns Hopkins University, B.H.P. is entitled to a share of royalties received by the University on sales of products. The terms of this arrangement are being managed by the Johns Hopkins University, in accordance with its conflict of interest policies. D.F.H. has received clinical and laboratory research funding from Janssen, AstraZeneca, and Pfizer. The University of Michigan holds two patents regarding isolation and characterization of circulating tumor cells, one of which has been licensed to Janssen, naming D.F.H. as the inventor. D.F.H. also has stock options in two diagnostic companies: InBiomotion and OncImmune. None of these conflicts pertains to this manuscript. All other authors declare no potential conflicts., (©2015 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
30. Ki-67 is required for maintenance of cancer stem cells but not cell proliferation.
- Author
-
Cidado J, Wong HY, Rosen DM, Cimino-Mathews A, Garay JP, Fessler AG, Rasheed ZA, Hicks J, Cochran RL, Croessmann S, Zabransky DJ, Mohseni M, Beaver JA, Chu D, Cravero K, Christenson ES, Medford A, Mattox A, De Marzo AM, Argani P, Chawla A, Hurley PJ, Lauring J, and Park BH
- Subjects
- Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Line, Tumor, Colonic Neoplasms genetics, Colonic Neoplasms pathology, Humans, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Signal Transduction, Breast Neoplasms metabolism, Colonic Neoplasms metabolism, Ki-67 Antigen metabolism
- Abstract
Ki-67 expression is correlated with cell proliferation and is a prognostic marker for various cancers; however, its function is unknown. Here we demonstrate that genetic disruption of Ki-67 in human epithelial breast and colon cancer cells depletes the cancer stem cell niche. Ki-67 null cells had a proliferative disadvantage compared to wildtype controls in colony formation assays and displayed increased sensitivity to various chemotherapies. Ki-67 null cancer cells showed decreased and delayed tumor formation in xenograft assays, which was associated with a reduction in cancer stem cell markers. Immunohistochemical analyses of human breast cancers revealed that Ki-67 expression is maintained at equivalent or greater levels in metastatic sites of disease compared to matched primary tumors, suggesting that maintenance of Ki-67 expression is associated with metastatic/clonogenic potential. These results elucidate Ki-67's role in maintaining the cancer stem cell niche, which has potential diagnostic and therapeutic implications for human malignancies.
- Published
- 2016
- Full Text
- View/download PDF
31. Familial GI Stromal Tumor With Loss of Heterozygosity and Amplification of Mutant KIT.
- Author
-
Forde PM, Cochran RL, Boikos SA, Zabransky DJ, Beaver JA, Meyer CF, Thornton KA, Montgomery EA, Lidor AO, Donehower RC, and Park BH
- Subjects
- Base Sequence, Female, Gastrointestinal Stromal Tumors enzymology, Gene Amplification, Humans, Loss of Heterozygosity, Middle Aged, Mutation, Pedigree, Gastrointestinal Neoplasms genetics, Gastrointestinal Stromal Tumors genetics, Proto-Oncogene Proteins c-kit genetics
- Published
- 2016
- Full Text
- View/download PDF
32. TMSB4Y is a candidate tumor suppressor on the Y chromosome and is deleted in male breast cancer.
- Author
-
Wong HY, Wang GM, Croessmann S, Zabransky DJ, Chu D, Garay JP, Cidado J, Cochran RL, Beaver JA, Aggarwal A, Liu ML, Argani P, Meeker A, Hurley PJ, Lauring J, and Park BH
- Subjects
- Actins genetics, Actins metabolism, Breast Neoplasms, Male metabolism, Breast Neoplasms, Male pathology, Cell Line, Cell Proliferation, Cell Shape, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Cell Transformation, Neoplastic pathology, Female, Gene Expression Regulation, Neoplastic, Humans, In Situ Hybridization, Fluorescence, Male, Mammary Glands, Human metabolism, Mammary Glands, Human pathology, Phenotype, Polymerase Chain Reaction, Thymosin metabolism, Time Factors, Transfection, Tumor Suppressor Proteins metabolism, Breast Neoplasms, Male genetics, Chromosomes, Human, Y, Gene Deletion, Thymosin genetics, Tumor Suppressor Proteins genetics
- Abstract
Male breast cancer comprises less than 1% of breast cancer diagnoses. Although estrogen exposure has been causally linked to the development of female breast cancers, the etiology of male breast cancer is unclear. Here, we show via fluorescence in situ hybridization (FISH) and droplet digital PCR (ddPCR) that the Y chromosome was clonally lost at a frequency of ~16% (5/31) in two independent cohorts of male breast cancer patients. We also show somatic loss of the Y chromosome gene TMSB4Y in a male breast tumor, confirming prior reports of loss at this locus in male breast cancers. To further understand the function of TMSB4Y, we created inducible cell lines of TMSB4Y in the female human breast epithelial cell line MCF-10A. Expression of TMSB4Y resulted in aberrant cellular morphology and reduced cell proliferation, with a corresponding reduction in the fraction of metaphase cells. We further show that TMSB4Y interacts directly with β-actin, the main component of the actin cytoskeleton and a cell cycle modulator. Taken together, our results suggest that clonal loss of the Y chromosome may contribute to male breast carcinogenesis, and that the TMSB4Y gene has tumor suppressor properties.
- Published
- 2015
- Full Text
- View/download PDF
33. HER2 missense mutations have distinct effects on oncogenic signaling and migration.
- Author
-
Zabransky DJ, Yankaskas CL, Cochran RL, Wong HY, Croessmann S, Chu D, Kavuri SM, Red Brewer M, Rosen DM, Dalton WB, Cimino-Mathews A, Cravero K, Button B, Kyker-Snowman K, Cidado J, Erlanger B, Parsons HA, Manto KM, Bose R, Lauring J, Arteaga CL, Konstantopoulos K, and Park BH
- Subjects
- Blotting, Western, Cell Line, Tumor, Cell Proliferation physiology, Colony-Forming Units Assay, Flow Cytometry, Gene Targeting, HEK293 Cells, Humans, Immunoblotting, Lapatinib, Quinazolines, Quinolines, Thiazoles, Cell Movement genetics, Mutation, Missense genetics, Neoplasms genetics, Receptor, ErbB-2 genetics, Signal Transduction genetics
- Abstract
Recurrent human epidermal growth factor receptor 2 (HER2) missense mutations have been reported in human cancers. These mutations occur primarily in the absence of HER2 gene amplification such that most HER2-mutant tumors are classified as "negative" by FISH or immunohistochemistry assays. It remains unclear whether nonamplified HER2 missense mutations are oncogenic and whether they are targets for HER2-directed therapies that are currently approved for the treatment of HER2 gene-amplified breast cancers. Here we functionally characterize HER2 kinase and extracellular domain mutations through gene editing of the endogenous loci in HER2 nonamplified human breast epithelial cells. In in vitro and in vivo assays, the majority of HER2 missense mutations do not impart detectable oncogenic changes. However, the HER2 V777L mutation increased biochemical pathway activation and, in the context of a PIK3CA mutation, enhanced migratory features in vitro. However, the V777L mutation did not alter in vivo tumorigenicity or sensitivity to HER2-directed therapies in proliferation assays. Our results suggest the oncogenicity and potential targeting of HER2 missense mutations should be considered in the context of cooperating genetic alterations and provide previously unidentified insights into functional analysis of HER2 mutations and strategies to target them.
- Published
- 2015
- Full Text
- View/download PDF
34. Comparison of cell stabilizing blood collection tubes for circulating plasma tumor DNA.
- Author
-
Toro PV, Erlanger B, Beaver JA, Cochran RL, VanDenBerg DA, Yakim E, Cravero K, Chu D, Zabransky DJ, Wong HY, Croessmann S, Parsons H, Hurley PJ, Lauring J, and Park BH
- Subjects
- Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Blood Chemical Analysis, Breast Neoplasms metabolism, Cancer Care Facilities, Class I Phosphatidylinositol 3-Kinases, Female, Hemolysis, Humans, Microchemistry methods, Mutation, Neoplasm Metastasis, Phosphatidylinositol 3-Kinases blood, Phosphatidylinositol 3-Kinases genetics, Phosphatidylinositol 3-Kinases metabolism, Plasma chemistry, Polymerase Chain Reaction, Prospective Studies, Signal Processing, Computer-Assisted, Biomarkers, Tumor blood, Breast Neoplasms blood, DNA, Neoplasm blood, Phlebotomy instrumentation
- Abstract
Objectives: Circulating plasma DNA is being increasingly used for biomedical and clinical research as a substrate for genetic testing. However, cell lysis can occur hours after venipuncture when using standard tubes for blood collection, leading to an increase in contaminating cellular DNA that may hinder analysis of circulating plasma DNA. Cell stabilization agents can prevent cellular lysis for several days, reducing the need for immediate plasma preparation after venipuncture, thereby facilitating the ease of blood collection and sample preparation for clinical research. However, the majority of cell stabilizing reagents have not been formally tested for their ability to preserve circulating plasma tumor DNA., Design & Methods: In this study, we compared the properties of two cell stabilizing reagents, the cell-free DNA BCT tube and the PAXgene tube, by collecting blood samples from metastatic breast cancer patients and measuring genome equivalents of plasma DNA by droplet digital PCR. We compared wild type PIK3CA genome equivalents and also assayed for two PIK3CA hotspot mutations, E545K and H1047R., Results: Our results demonstrate that blood stored for 7 days in BCT tubes did not show evidence of cell lysis, whereas PAXgene tubes showed an order of magnitude increase in genome equivalents, indicative of considerable cellular lysis., Conclusions: We conclude that BCT tubes can prevent lysis and cellular release of genomic DNA of blood samples from cancer patients when stored at room temperature, and could therefore be of benefit for blood specimen collections in clinical trials., (Copyright © 2015 The Canadian Society of Clinical Chemists. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
35. Functional isogenic modeling of BRCA1 alleles reveals distinct carrier phenotypes.
- Author
-
Cochran RL, Cidado J, Kim M, Zabransky DJ, Croessmann S, Chu D, Wong HY, Beaver JA, Cravero K, Erlanger B, Parsons H, Heaphy CM, Meeker AK, Lauring J, and Park BH
- Subjects
- Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Cycle radiation effects, Cell Line, Transformed, Cell Proliferation radiation effects, Centrosome metabolism, Centrosome radiation effects, Female, Gene Expression Regulation, Neoplastic, Genetic Predisposition to Disease, Genomic Instability, Heredity, Heterozygote, Humans, Phenotype, Radiation Tolerance, Risk Assessment, Risk Factors, Time Factors, Transfection, BRCA1 Protein genetics, Biomarkers, Tumor genetics, Breast Neoplasms genetics, Models, Genetic, Mutation
- Abstract
Clinical genetic testing of BRCA1 and BRCA2 is commonly performed to identify specific individuals at risk for breast and ovarian cancers who may benefit from prophylactic therapeutic interventions. Unfortunately, it is evident that deleterious BRCA1 alleles demonstrate variable penetrance and that many BRCA1 variants of unknown significance (VUS) exist. In order to further refine hereditary risks that may be associated with specific BRCA1 alleles, we performed gene targeting to establish an isogenic panel of immortalized human breast epithelial cells harboring eight clinically relevant BRCA1 alleles. Interestingly, BRCA1 mutations and VUS had distinct, quantifiable phenotypes relative to isogenic parental BRCA1 wild type cells and controls. Heterozygous cells with known deleterious BRCA1 mutations (185delAG, C61G and R71G) demonstrated consistent phenotypes in radiation sensitivity and genomic instability assays, but showed variability in other assays. Heterozygous BRCA1 VUS cells also demonstrated assay variability, with some VUS demonstrating phenotypes more consistent with deleterious alleles. Taken together, our data suggest that BRCA1 deleterious mutations and VUS can differ in their range of tested phenotypes, suggesting they might impart varying degrees of risk. These results demonstrate that functional isogenic modeling of BRCA1 alleles could aid in classifying BRCA1 mutations and VUS, and determining BRCA allele cancer risk.
- Published
- 2015
- Full Text
- View/download PDF
36. NDRG1 links p53 with proliferation-mediated centrosome homeostasis and genome stability.
- Author
-
Croessmann S, Wong HY, Zabransky DJ, Chu D, Mendonca J, Sharma A, Mohseni M, Rosen DM, Scharpf RB, Cidado J, Cochran RL, Parsons HA, Dalton WB, Erlanger B, Button B, Cravero K, Kyker-Snowman K, Beaver JA, Kachhap S, Hurley PJ, Lauring J, and Park BH
- Subjects
- Aneuploidy, Animals, Breast metabolism, Cell Line, Cell Proliferation, Centrosome metabolism, Female, Genome, Heterozygote, Homeostasis, Homozygote, Humans, In Situ Hybridization, Fluorescence, Mice, Mice, Knockout, Neoplasms pathology, Phenotype, RNA Interference, Tubulin metabolism, Cell Cycle Proteins metabolism, Centrosome ultrastructure, Gene Expression Regulation, Neoplastic, Intracellular Signaling Peptides and Proteins metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
The tumor protein 53 (TP53) tumor suppressor gene is the most frequently somatically altered gene in human cancers. Here we show expression of N-Myc down-regulated gene 1 (NDRG1) is induced by p53 during physiologic low proliferative states, and mediates centrosome homeostasis, thus maintaining genome stability. When placed in physiologic low-proliferating conditions, human TP53 null cells fail to increase expression of NDRG1 compared with isogenic wild-type controls and TP53 R248W knockin cells. Overexpression and RNA interference studies demonstrate that NDRG1 regulates centrosome number and amplification. Mechanistically, NDRG1 physically associates with γ-tubulin, a key component of the centrosome, with reduced association in p53 null cells. Strikingly, TP53 homozygous loss was mutually exclusive of NDRG1 overexpression in over 96% of human cancers, supporting the broad applicability of these results. Our study elucidates a mechanism of how TP53 loss leads to abnormal centrosome numbers and genomic instability mediated by NDRG1.
- Published
- 2015
- Full Text
- View/download PDF
37. A phosphoproteomic screen demonstrates differential dependence on HER3 for MAP kinase pathway activation by distinct PIK3CA mutations.
- Author
-
Blair BG, Wu X, Zahari MS, Mohseni M, Cidado J, Wong HY, Beaver JA, Cochran RL, Zabransky DJ, Croessmann S, Chu D, Toro PV, Cravero K, Pandey A, and Park BH
- Subjects
- Breast Neoplasms genetics, Cell Line, Cell Line, Tumor, Cell Proliferation, Class I Phosphatidylinositol 3-Kinases, Female, Gene Expression Regulation, Neoplastic, Humans, Mutation, Phosphatidylinositol 3-Kinases chemistry, Phosphatidylinositol 3-Kinases genetics, Phosphorylation, Protein Structure, Tertiary, RNA Interference, Receptor, ErbB-3 genetics, Breast Neoplasms metabolism, Mitogen-Activated Protein Kinases metabolism, Phosphatidylinositol 3-Kinases metabolism, Proteomics, Receptor, ErbB-3 metabolism, Signal Transduction
- Abstract
The PIK3CA gene encodes for the p110 alpha isoform of PI3 kinase and is one of the most frequently mutated oncogenes in human cancers. However, the mechanisms by which PIK3CA mutations activate cell signaling are not fully understood. Here we used a phosphoproteomic approach to compare differential phosphorylation patterns between human breast epithelial cells and two isogenic somatic cell knock in derivatives, each harboring a distinct PIK3CA mutation. We demonstrated differential phosphorylation patterns between isogenic cell lines containing a PIK3CA helical domain mutation (E545K) compared to cells with a PIK3CA kinase domain mutation (H1047R). In particular, the receptor tyrosine kinase, HER3, showed increased phosphorylation at tyrosine 1328 in H1047R cells versus E545K cells. Genetic studies using shRNA demonstrated that H1047R cells have a profound decrease in growth factor independent proliferation upon HER3 knock down, but this effect was attenuated in E545K cells. In addition, HER3 knock down led to reductions in both PI3 kinase and MAP kinase pathway activation in H1047R cells, but in E545K cells only PI3 kinase pathway diminution was observed. These studies demonstrate the power of using paired isogenic cell lines for proteomic analysis to gain new insights into oncogenic signal transduction pathways., (© 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2015
- Full Text
- View/download PDF
38. MACROD2 overexpression mediates estrogen independent growth and tamoxifen resistance in breast cancers.
- Author
-
Mohseni M, Cidado J, Croessmann S, Cravero K, Cimino-Mathews A, Wong HY, Scharpf R, Zabransky DJ, Abukhdeir AM, Garay JP, Wang GM, Beaver JA, Cochran RL, Blair BG, Rosen DM, Erlanger B, Argani P, Hurley PJ, Lauring J, and Park BH
- Subjects
- Base Sequence, Cell Line, Tumor, Cell Proliferation, Epigenesis, Genetic, Female, Gene Deletion, Gene Dosage, Humans, Molecular Sequence Data, Neoplasm Transplantation, Phenotype, Prognosis, RNA Interference, RNA, Small Interfering metabolism, Receptors, Estrogen metabolism, Transgenes, Treatment Outcome, Breast Neoplasms metabolism, DNA Repair Enzymes metabolism, Drug Resistance, Neoplasm, Estrogens metabolism, Hydrolases metabolism, Tamoxifen pharmacology
- Abstract
Tamoxifen is effective for treating estrogen receptor-alpha (ER) positive breast cancers. However, few molecular mediators of tamoxifen resistance have been elucidated. Here we describe a previously unidentified gene, MACROD2 that confers tamoxifen resistance and estrogen independent growth. We found MACROD2 is amplified and overexpressed in metastatic tamoxifen-resistant tumors. Transgene overexpression of MACROD2 in breast cancer cell lines results in tamoxifen resistance, whereas RNAi-mediated gene knock down reverses this phenotype. MACROD2 overexpression also leads to estrogen independent growth in xenograft assays. Mechanistically, MACROD2 increases p300 binding to estrogen response elements in a subset of ER regulated genes. Primary breast cancers and matched metastases demonstrate MACROD2 expression can change with disease evolution, and increased expression and amplification of MACROD2 in primary tumors is associated with worse overall survival. These studies establish MACROD2 as a key mediator of estrogen independent growth and tamoxifen resistance, as well as a potential novel target for diagnostics and therapy.
- Published
- 2014
- Full Text
- View/download PDF
39. Analysis of BRCA2 loss of heterozygosity in tumor tissue using droplet digital polymerase chain reaction.
- Author
-
Cochran RL, Cravero K, Chu D, Erlanger B, Toro PV, Beaver JA, Zabransky DJ, Wong HY, Cidado J, Croessmann S, Parsons H, Kim M, Wheelan SJ, Argani P, and Ho Park B
- Subjects
- Alleles, Breast Neoplasms pathology, Carcinoma, Ductal, Breast pathology, Female, Genotype, Humans, Middle Aged, Mutation, BRCA2 Protein genetics, Breast Neoplasms genetics, Carcinoma, Ductal, Breast genetics, Loss of Heterozygosity, Polymerase Chain Reaction methods
- Abstract
Loss-of-heterozygosity (LOH) analysis of archival tumor tissue can aid in determining the clinical significance of BRCA variants. Here we describe an approach for assessing LOH in formalin-fixed, paraffin-embedded (FFPE) tissues using variant-specific probes and droplet digital polymerase chain reaction (ddPCR). We evaluated LOH in 2 related breast cancer patients harboring a rare missense BRCA2 variant of unknown clinical significance (c.6966G>T; M2322I). Conventional PCR followed by Sanger sequencing suggested a change in allelic abundance in the FFPE specimens. However, we found no evidence of LOH as determined by the allelic ratio (wild type-variant) for BRCA2 in both patients' archival tumor specimens and adjacent normal control tissues using ddPCR. In summary, these experiments demonstrate the utility of ddPCR to quickly and accurately assess LOH in archival FFPE tumor tissue., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
40. Detection of cancer DNA in plasma of patients with early-stage breast cancer.
- Author
-
Beaver JA, Jelovac D, Balukrishna S, Cochran R, Croessmann S, Zabransky DJ, Wong HY, Toro PV, Cidado J, Blair BG, Chu D, Burns T, Higgins MJ, Stearns V, Jacobs L, Habibi M, Lange J, Hurley PJ, Lauring J, VanDenBerg D, Kessler J, Jeter S, Samuels ML, Maar D, Cope L, Cimino-Mathews A, Argani P, Wolff AC, and Park BH
- Subjects
- Adult, Aged, Breast Neoplasms surgery, Class I Phosphatidylinositol 3-Kinases, DNA Mutational Analysis methods, DNA, Neoplasm chemistry, Exons genetics, Female, Humans, Middle Aged, Mutation, Neoplasm Staging, Phosphatidylinositol 3-Kinases genetics, Polymerase Chain Reaction methods, Postoperative Period, Preoperative Period, Prospective Studies, Reproducibility of Results, Sensitivity and Specificity, Breast Neoplasms diagnosis, Breast Neoplasms genetics, DNA, Neoplasm blood, DNA, Neoplasm genetics
- Abstract
Purpose: Detecting circulating plasma tumor DNA (ptDNA) in patients with early-stage cancer has the potential to change how oncologists recommend systemic therapies for solid tumors after surgery. Droplet digital polymerase chain reaction (ddPCR) is a novel sensitive and specific platform for mutation detection., Experimental Design: In this prospective study, primary breast tumors and matched pre- and postsurgery blood samples were collected from patients with early-stage breast cancer (n = 29). Tumors (n = 30) were analyzed by Sanger sequencing for common PIK3CA mutations, and DNA from these tumors and matched plasma were then analyzed for PIK3CA mutations using ddPCR., Results: Sequencing of tumors identified seven PIK3CA exon 20 mutations (H1047R) and three exon 9 mutations (E545K). Analysis of tumors by ddPCR confirmed these mutations and identified five additional mutations. Presurgery plasma samples (n = 29) were then analyzed for PIK3CA mutations using ddPCR. Of the 15 PIK3CA mutations detected in tumors by ddPCR, 14 of the corresponding mutations were detected in presurgical ptDNA, whereas no mutations were found in plasma from patients with PIK3CA wild-type tumors (sensitivity 93.3%, specificity 100%). Ten patients with mutation-positive ptDNA presurgery had ddPCR analysis of postsurgery plasma, with five patients having detectable ptDNA postsurgery., Conclusions: This prospective study demonstrates accurate mutation detection in tumor tissues using ddPCR, and that ptDNA can be detected in blood before and after surgery in patients with early-stage breast cancer. Future studies can now address whether ptDNA detected after surgery identifies patients at risk for recurrence, which could guide chemotherapy decisions for individual patients., (©2014 American Association for Cancer Research.)
- Published
- 2014
- Full Text
- View/download PDF
41. Estrogen receptor and receptor tyrosine kinase signaling: use of combinatorial hormone and epidermal growth factor receptor/human epidermal growth factor receptor 2-targeted therapies for breast cancer.
- Author
-
Zabransky DJ and Park BH
- Subjects
- Antineoplastic Agents, Hormonal therapeutic use, Class I Phosphatidylinositol 3-Kinases, Female, Humans, Molecular Targeted Therapy, Phosphatidylinositol 3-Kinases genetics, Phosphatidylinositol 3-Kinases metabolism, Signal Transduction, Breast Neoplasms drug therapy, ErbB Receptors antagonists & inhibitors, Protein-Tyrosine Kinases metabolism, Receptor, ErbB-2 antagonists & inhibitors, Receptors, Estrogen metabolism
- Published
- 2014
- Full Text
- View/download PDF
42. Lenalidomide modulates IL-8 and anti-prostate antibody levels in men with biochemically recurrent prostate cancer.
- Author
-
Zabransky DJ, Smith HA, Thoburn CJ, Zahurak M, Keizman D, Carducci M, Eisenberger MA, McNeel DG, Drake CG, and Antonarakis ES
- Subjects
- Adenocarcinoma diagnosis, Adenocarcinoma immunology, Aged, Aged, 80 and over, Antigens, Surface immunology, Biomarkers, Tumor immunology, Disease Progression, Humans, Immunomodulation drug effects, Lenalidomide, Male, Middle Aged, Neoplasm Recurrence, Local diagnosis, Neoplasm Recurrence, Local immunology, Prostate-Specific Antigen blood, Prostatectomy, Prostatic Neoplasms diagnosis, Prostatic Neoplasms immunology, Radiotherapy, Adjuvant, Retrospective Studies, Thalidomide therapeutic use, Adenocarcinoma therapy, Antineoplastic Agents therapeutic use, Interleukin-8 metabolism, Neoplasm Recurrence, Local drug therapy, Prostate-Specific Antigen immunology, Prostatic Neoplasms therapy, Thalidomide analogs & derivatives
- Abstract
Background: We retrospectively explored changes in immunological parameters in men with biochemically recurrent prostate cancer treated with either 5 or 25 mg of lenalidomide in a randomized phase 2 trial, and determined whether those changes correlated with disease progression., Methods: Cytokine levels were compared for each patient at baseline and after 6 months of treatment with lenalidomide. Regression models for correlated data were used to assess associations of cytokine levels with lenalidomide treatment effect. Estimates were obtained using generalized estimating equations. Changes in circulating anti-prostate antibodies were evaluated using a high-throughput immunoblot technique., Results: Treatment with lenalidomide was associated with global changes in immunoreactivity to a number of prostate-associated antigens, as well as with changes in circulating levels of the T(H) 2 cytokines IL-4, IL-5, IL-10, and IL-13. Disease progression in treated patients was associated with an increase in circulating IL-8 levels, while IL-8 levels decreased significantly in non-progressors., Conclusions: Lenalidomide demonstrates immunomodulatory properties in patients with biochemically recurrent prostate cancer. The induction of novel anti-prostate antibodies is a potential mechanism for lenalidomide response. Changes in serum IL-8 levels may serve as a potential biomarker in treated patients. These hypotheses require formal testing in future prospective trials., (Copyright © 2011 Wiley Periodicals, Inc.)
- Published
- 2012
- Full Text
- View/download PDF
43. Phenotypic and functional properties of Helios+ regulatory T cells.
- Author
-
Zabransky DJ, Nirschl CJ, Durham NM, Park BV, Ceccato CM, Bruno TC, Tam AJ, Getnet D, and Drake CG
- Subjects
- Animals, Antigens, CD genetics, Gene Expression Regulation, Neoplastic, Glucocorticoid-Induced TNFR-Related Protein genetics, Integrin alpha Chains genetics, Mice, Mice, Inbred BALB C, Neoplasms genetics, Phenotype, Spleen cytology, T-Lymphocytes, Regulatory cytology, Up-Regulation, DNA-Binding Proteins genetics, T-Lymphocytes, Regulatory metabolism, Transcription Factors genetics
- Abstract
Helios, an Ikaros family transcription factor, is preferentially expressed at the mRNA and protein level in regulatory T cells. Helios expression previously appeared to be restricted to thymic-derived Treg. Consistent with recent data, we show here that Helios expression is inducible in vitro under certain conditions. To understand phenotypic and functional differences between Helios(+) and Helios(-) Treg, we profiled cell-surface markers of FoxP3(+) Treg using unmanipulated splenocytes. We found that CD103 and GITR are expressed at high levels on a subset of Helios(+) Treg and that a Helios(+) Treg population could be significantly enriched by FACS sorting using these two markers. Quantitative real-time PCR (qPCR) analysis revealed increased TGF-β message in Helios(+) Treg, consistent with the possibility that this population possesses enhanced regulatory potential. In tumor-bearing mice, we found that Helios(+) Treg were relatively over-represented in the tumor-mass, and BrdU studies showed that, in vivo, Helios(+) Treg proliferated more than Helios(-) Treg. We hypothesized that Helios-enriched Treg might exert increased suppressive effects. Using in vitro suppression assays, we show that Treg function correlates with the absolute number of Helios(+) cells in culture. Taken together, these data show that Helios(+) Treg represent a functional subset with associated CD103 and GITR expression.
- Published
- 2012
- Full Text
- View/download PDF
44. Electron tomography of the contact between T cells and SIV/HIV-1: implications for viral entry.
- Author
-
Sougrat R, Bartesaghi A, Lifson JD, Bennett AE, Bess JW, Zabransky DJ, and Subramaniam S
- Subjects
- Binding Sites, CD4 Antigens metabolism, CD4 Antigens physiology, HIV Envelope Protein gp120 metabolism, HIV Envelope Protein gp120 physiology, HIV Envelope Protein gp41 metabolism, HIV Envelope Protein gp41 physiology, Imaging, Three-Dimensional, Virion, CD4-Positive T-Lymphocytes virology, Electrons, HIV-1 pathogenicity, Simian Immunodeficiency Virus pathogenicity, Tomography methods
- Abstract
The envelope glycoproteins of primate lentiviruses, including human and simian immunodeficiency viruses (HIV and SIV), are heterodimers of a transmembrane glycoprotein (usually gp41), and a surface glycoprotein (gp120), which binds CD4 on target cells to initiate viral entry. We have used electron tomography to determine the three-dimensional architectures of purified SIV virions in isolation and in contact with CD4+ target cells. The trimeric viral envelope glycoprotein surface spikes are heterogeneous in appearance and typically approximately 120 A long and approximately 120 A wide at the distal end. Docking of SIV or HIV-1 on the T cell surface occurs via a neck-shaped contact region that is approximately 400 A wide and consistently consists of a closely spaced cluster of five to seven rod-shaped features, each approximately 100 A long and approximately 100 A wide. This distinctive structure is not observed when viruses are incubated with T lymphocytes in the presence of anti-CD4 antibodies, the CCR5 antagonist TAK779, or the peptide entry inhibitor SIVmac251 C34. For virions bound to cells, few trimers were observed away from this cluster at the virion-cell interface, even in cases where virus preparations showing as many as 70 envelope glycoprotein trimers per virus particle were used. This contact zone, which we term the "entry claw", provides a spatial context to understand the molecular mechanisms of viral entry. Determination of the molecular composition and structure of the entry claw may facilitate the identification of improved drugs for the inhibition of HIV-1 entry.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.