scispace - formally typeset
Search or ask a question

Showing papers by "Joan S. Brugge published in 2022"


Journal ArticleDOI
TL;DR: In this paper , the authors describe mechanisms by which tumour ecosystems, including cancer cells, immune cells and stroma, adapt to therapeutic stresses and describe three different approaches to exploit stress mitigation processes: (1) interdict stress mitigation to induce cell death; (2) increase stress to induce cellular catastrophe; and (3) exploit emergent vulnerabilities in cancer cells and cells of the tumour microenvironment.
Abstract: Normal cells explore multiple states to survive stresses encountered during development and self-renewal as well as environmental stresses such as starvation, DNA damage, toxins or infection. Cancer cells co-opt normal stress mitigation pathways to survive stresses that accompany tumour initiation, progression, metastasis and immune evasion. Cancer therapies accentuate cancer cell stresses and invoke rapid non-genomic stress mitigation processes that maintain cell viability and thus represent key targetable resistance mechanisms. In this Review, we describe mechanisms by which tumour ecosystems, including cancer cells, immune cells and stroma, adapt to therapeutic stresses and describe three different approaches to exploit stress mitigation processes: (1) interdict stress mitigation to induce cell death; (2) increase stress to induce cellular catastrophe; and (3) exploit emergent vulnerabilities in cancer cells and cells of the tumour microenvironment. We review challenges associated with tumour heterogeneity, prioritizing actionable adaptive responses for optimal therapeutic outcomes, and development of an integrative framework to identify and target vulnerabilities that arise from adaptive responses and engagement of stress mitigation pathways. Finally, we discuss the need to monitor adaptive responses across multiple scales and translation of combination therapies designed to take advantage of adaptive responses and stress mitigation pathways to the clinic.

56 citations


Journal ArticleDOI
TL;DR: In this article , a unified, high-resolution breast atlas is presented by integrating single-cell RNA-seq, mass cytometry, and cyclic immunofluorescence, encompassing a myriad of states.

19 citations


Journal ArticleDOI
TL;DR: Treatment of lung cancer with EGFR inhibitors with AXL overexpression cured patient-derived xenografts and uncovered another hypermutator, AXL-driven activation of MYC and increased purine synthesis that is unbalanced by pyrimidines.
Abstract: Resistance of EGFR-mutant lung cancers to EGFR inhibitors can occur through engagement of mutation-prone DNA replication activated by GAS6 and AXL upregulation, and blocking this pathway can prevent resistance and offer new treatment strategies.

10 citations


Journal ArticleDOI
TL;DR: McDermott et al. as mentioned in this paper developed an anti-CLDN6 antibody drug conjugate for the treatment of CLDN6 positive cancers, which is currently in Phase I clinical testing.
Abstract: Background: Claudin 6 (CLDN6), a member of the claudin family of tight junction proteins, is expressed at high levels in multiple human malignancies including ovarian and endometrial cancers. Conversely it has little or no expression in normal tissues. This expression profile makes CLDN6 an ideal target for development of potential therapeutic antibody-drug conjugates (ADCs). This study describes the generation and preclinical characterization of an anti-CLDN6 ADC consisting of a humanized anti-CLDN6 monoclonal antibody coupled to MMAE via a cleavable linker. Materials and Methods: A fully humanized anti-CLDN6 antibody was initially characterized for binding affinity, selectivity/specificity, internalization characteristics and in vivo efficacy. It was then conjugated to MMAE resulting in the potential therapeutic anti-CLDN6 ADC. The anti-tumor efficacy of the ADC was next assessed for anti-tumor efficacy in CLDN6 positive (CLDN6+) and negative (CLDN6-) xenografts and patient-derived xenograft (PDX) models of specific cancers including ovarian and endometrial cancer. Results: Selective binding of the ADC to CLDN6, without cross reactivity to other CLDN family members CLDN3, CLDN4 and CLDN9, was confirmed in human cancer cell lines and cells engineered to overexpress each protein. The ADC was also shown to rapidly internalize in CLDN6+ cells. Robust tumor regressions following treatment with the ADC were observed in CLDN6+ xenografts that were sustained beyond the treatment window. Conversely, there was limited to no activity of the ADC in CLDN6- xenografts models. In addition, the prevalence of CLDN6 expression in human ovarian and endometrial cancers was assessed by IHC in tissue microarrays and found to be 28% (ovarian epithelial carcinomas) and 11% (endometrial carcinomas), respectively. Discussion: Overall, these data suggest that our anti-CLDN6 ADC may be a promising treatment for patients with CLDN6+ tumors and it is currently in Phase I clinical testing. Citation Format: Martina S. McDermott, Ke Wei Gong, Neil A. O'Brien, Dylan Conklin, Benjamin Hoffstrom, Ming Lu, Jun Zhang, Tong Luo, Weiping Jia, Jenny J. Hong, Kevin Chau, Simon Davenport, Michael F. Press, Abram Handly-Santana, Joan S. Brugge, Ronny Drapkin, John A. Glaspy, Leonard Presta, Dennis J. Slamon. Development and characterization of a novel anti-CLDN6 antibody drug conjugate for the treatment of CLDN6 positive cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 342.

1 citations


Journal ArticleDOI
TL;DR: A Phase 1 trial of the MEK inhibitor trametinib with the BCLxL inhibitor navitoclax was conducted to establish the recommended phase 2 dose (RP2D) and activity in select RAS-mutated GYN cancers as mentioned in this paper .

1 citations


Journal ArticleDOI
TL;DR: The mouse model of AI and GI shows that suppression of estrogen signaling after initiation of AI offered marked protection against precancerous changes, providing a rationale for considering short-term TAM treatment for women who do not breastfeed to reduce risk of BC.
Abstract: Background: Triple negative breast cancer (TNBC) is associated with poor survival, particularly affecting African American women (AAW). Epidemiological studies indicate prolonged breast feeding reduces breast cancer (BC) risk, including TNBC. AAW have significantly lower rates of breast feeding compared to Caucasian women. To understand this link we developed a mouse model mimicking abrupt (AI) and gradual involution (GI). AI led to increased estrogen signaling, cell proliferation and chronic inflammation, which was followed by hyperplasia and squamous metaplasia in mammary glands1. There was an increase in the luminal progenitor (LP) cell population, the cells of origin of TNBC, and a decrease in mature luminal (ML) cells in AI glands. In this study, we sought to determine if blocking estrogen signaling with tamoxifen (TAM) could revert the negative effects of AI, and if so, could be a prophylactic option to reduce BC risk in women who do not breast feed. Methods: Uniparous FVB/N mice (~8 weeks) were allowed to nurse six pups per dam at partum. To induce AI, all pups were removed on postpartum (PP) day 7 (d7). For TAM treatment, 5mg sustained release TAM citrate pellet or placebo was implanted in the subscapular region on PP d8. Mammary glands were harvested on PP d28 and d120. FFPE sections were used for histology and immunohistochemistry. Single cell suspensions were analyzed for mammary epithelial subpopulations using Fluorescence Activated Cell Sorting. Affymetrix and qPCR were used for gene expression analysis. Mass cytometry was performed on mammary glands harvested at PP d120. Results: TAM treatment for 21 days completely abrogated hyperplastic and metaplastic changes in AI glands harvested on d120. Treatment initiation on PP d8, d15 and d35 had the same effect. TAM treatment reduced the cell proliferation and collagen deposition in AI glands. De-enrichment of estrogen signaling pathways and decrease in Elf5 expression, a luminal progenitor marker, were observed upon TAM treatment in d28 glands. Mass cytometry revealed a marked reduction in LP population and a significant increase in ML population in TAM treated AI glands on d120, restoring to the levels in age matched virgin mice. Significant increases in progenitor-like markers TSPAN8, Ly6D, CD200 and decreases in CD49f and CD47 expression in LP cells were observed, indicating return to a normal uniparous LP state. Expression of Ly-6D in ML cells, a ML cell marker, was also rescued upon TAM treatment. Conclusion: Using our mouse model of AI and GI, we show that suppression of estrogen signaling after initiation of AI offered marked protection against precancerous changes. TAM restored the balance of epithelial lineages and normalized the LP and basal cells in AI glands to the post-involution phenotype. Our data provide a rationale for considering short-term TAM treatment for women who do not breastfeed to reduce risk of BC. 1. Basree et.al. PMID 31315645 Citation Format: Bhuvaneswari Ramaswamy, Neelam Shinde, Gary K. Gray, Resham Mawalkar, Allen Zhang, Mustafa Basree, Xiaoli Zhang, Ramesh Ganju, Gina M. Sizemore, Joan S. Brugge, Sarmila Majumder. Prophylactic use of tamoxifen could reduce the risk of breast cancer in women who do not breast feed postpartum [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 11.

Posted ContentDOI
03 Nov 2022-bioRxiv
TL;DR: In this article , the BRCA1-IRIS (IRIS) isoform has critical functions in the Fanconi anemia (FA) pathway and in repair of DNA interstrand crosslinks.
Abstract: The tumor suppressor BRCA1 encodes multiple protein products including the canonical BRCA1-p220 (p220), which plays important roles in repair of diverse types of DNA damage. However, contributions of other BRCA1-encoded protein isoforms to DNA damage repair are less clear. Here, we report that the BRCA1-IRIS (IRIS) isoform has critical functions in the Fanconi anemia (FA) pathway and in repair of DNA interstrand crosslinks (ICLs). Loss of IRIS expression sensitizes cells to ICLs and impairs ICL repair. ICL formation stimulates association of IRIS with both FANCD2 and the FA core complex, which promotes FANCD2 recruitment to damage sites. The unique, BRCA1 intron 11-encoded C-terminal tail of IRIS is required for complex formation with FANCD2 and for ICL-inducible FANCD2 mono-ubiquitylation. Collectively, our findings reveal that IRIS plays an essential role, upstream of the p220-directed HR, in the FA pathway through a previously unrecognized mechanism that depends on the IRIS-FANCA-FANCD2 interaction. Highlights BRCA1 splicing isoform BRCA1-IRIS is required for interstrand crosslink (ICL) repair. BRCA1-IRIS interacts with FANCD2 and promotes its recruitment to sites of ICL damage. BRCA1-IRIS, but not BRCA1-p220, promotes ICL-inducible FANCD2 mono-ubiquitylation. The unique C-terminal tail of BRCA1-IRIS is essential for its function in ICL repair.

Journal ArticleDOI
TL;DR: The integrative approach reveals that B1 heterozygosity in combination with RS leads to accumulation and proliferation of a specific mammary cell population that contributes to breast tumorigenesis, and scRNAseq-based analysis has revealed early changes that occur in the breast tissue as different cell types respond to RS.
Abstract: Women with BRCA1 (B1) mutation have an exceptionally high risk of developing breast cancer. The only effective preventive strategy currently offered to these women is the life altering prophylactic mastectomy. In light of limited treatment options available, it is critical that new therapeutic and preventive strategies be identified. Design of such strategies requires an understanding of early events in the breast cells that drive tumorigenesis. B1 heterozygous mouse models can help us identify these early changes in mammary tissue as the cells become tumor cells. However, despite the well-established association between B1 heterozygosity and cancer predisposition in humans, there are currently no such B1 mouse models that faithfully recapitulate this high risk of tumor formation. B1 heterozygous mice are not tumor-prone. This makes it difficult to use these models to study the role of B1 heterozygosity and to identify early tumor promoting changes in the breast tissue. We have now established a mouse model that induces mammary tumors in B1 heterozygous (Brca1wt/flx,Trp53flx/flx,K14cre) mice upon replication stress (RS), thus giving us a tool to study early tumor promoting changes in B1 heterozygous breast tissue. Our approach is based on our published work that reveals haploinsufficiency for RS suppression in B1 heterozygous cells. Given the importance of RS development in tumorigenesis, this effect would be a logical contributor to B1 mutant cancer development. Indeed, increasing RS in B1 heterozygous mice resulted in accelerated mammary tumorigenesis. RS in this mouse model was delivered by injecting DNA-adduct forming 4-nitroquinoline-1-oxide via mammary intraductal injection over a course of 7 weeks. RS served as an efficient and abnormally rapid driver of tumor formation (30 days post completion of injection regimen) in B1 heterozygous, but not B1 wild type mice. B1 heterozygous mice formed mammary adenocarcinoma and ductal carcinoma in-situ. Immunofluorescence based tissue section analysis and transcriptomic analysis reveals that adenocarcinomas formed in B1 heterozygous mice carry a basal epithelial phenotype like those found in human breast cancer. Such an accelerated tumor model system could prove to be invaluable in understanding the earliest events in B1 mutant breast cancer.Furthermore, our scRNAseq-based analysis has revealed early changes that occur in the breast tissue as different cell types (luminal and basal) respond to RS, and have identified new cell populations that emerge exclusively in B1 heterozygous mammary tissue undergoing RS. For this analysis, cells were collected from naïve mammary tissue, tissue collected midway through injections, and post-tumor tissue. This analysis identified a unique population of trans-differentiated cells expressing prognostic markers that have correlation to poor outcome in human breast cancer. This RS-induced mammary cell population in B1 heterozygous tissue also expresses both luminal progenitor and basal epithelial markers. Interestingly, this population was enriched for proliferation markers like Top2a, Ube2c, mKi67, and Ccnb2. Given that such proliferation markers are a hallmark of cancer stem cells, we suspect that this transdifferentiated population, which is primarily enriched in B1 heterozygous mammary tissue undergoing RS, marks some of the early cancer promoting changes in the breast tissue.Altogether, our integrative approach reveals that B1 heterozygosity in combination with RS leads to accumulation and proliferation of a specific mammary cell population that contributes to breast tumorigenesis. Identification of such early drivers is critical for the design of effective preventive and therapeutic strategies for women with B1 mutation. Citation Format: Shailja Pathania, Joshua Rivera, Delan Khalid, Monica Manne, Stevenson Tran, Kemmie Kibaja, Carman MC Li, Joan Brugge. Single cell RNA transcriptomics reveals tumor promoting mammary cell subpopulation upon replication stress in BRCA1 mutant breast cancer mouse model [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr P5-01-02.

Journal ArticleDOI
TL;DR: In this article , layer-by-layer nanoparticles (LbL NPs) are used to co-encapulate BCL2 and BCL-XL inhibitors in order to target HGSOC tumor cells and reduce systemic toxicities.
Abstract: Abstract The majority of patients with high grade serous ovarian cancer (HGSOC) develop recurrent disease and chemotherapy resistance. To identify drug combinations that would be effective in treatment of chemotherapy resistant disease, we examined the efficacy of drug combinations that target the three antiapoptotic proteins most commonly expressed in HGSOC—BCL2, BCL‐XL, and MCL1. Co‐inhibition of BCL2 and BCL‐XL (ABT‐263) with inhibition of MCL1 (S63845) induces potent synergistic cytotoxicity in multiple HGSOC models. Since this drug combination is predicted to be toxic to patients due to the known clinical morbidities of each drug, we developed layer‐by‐layer nanoparticles (LbL NPs) that co‐encapsulate these inhibitors in order to target HGSOC tumor cells and reduce systemic toxicities. We show that the LbL NPs can be designed to have high association with specific ovarian tumor cell types targeted in these studies, thus enabling a more selective uptake when delivered via intraperitoneal injection. Treatment with these LbL NPs displayed better potency than free drugs in vitro and resulted in near‐complete elimination of solid tumor metastases of ovarian cancer xenografts. Thus, these results support the exploration of LbL NPs as a strategy to deliver potent drug combinations to recurrent HGSOC. While these findings are described for co‐encapsulation of a BCL2/XL and a MCL1 inhibitor, the modular nature of LbL assembly provides flexibility in the range of therapies that can be incorporated, making LbL NPs an adaptable vehicle for delivery of additional combinations of pathway inhibitors and other oncology drugs.

Journal ArticleDOI
TL;DR: In this article , the G2 glyco-protein was identified and mapped using recombinant Escherichia coli expression clones and the M segment proteins involved in viral entry using in vitro transcription/transla-tion and recombinant vaccinia viruses.
Abstract: characterized its mRNA (12). They elucidated the biogenesis and subcellular localization of the M segment proteins involved in viral entry using in vitro transcription/transla-tion and recombinant vaccinia viruses (13 – 16). Antigenic determinants on the G2 glyco-protein were identi fi ed and mapped using recombinant Escherichia coli expression clones (17). Marc ’ s contributions in this area are still cited today in publications on RVFV vaccine development.

Posted ContentDOI
16 Nov 2022-bioRxiv
TL;DR: In this paper , the authors report that BRCA2 mutation carriers preferentially develop luminal-like breast cancers, but it remains unclear how BRCa2 mutations affect mammary epithelial subpopulations.
Abstract: BRCA2 mutation carriers preferentially develop luminal-like breast cancers, but it remains unclear how BRCA2 mutations affect mammary epithelial subpopulations. Here, we report that Brca2mut/WT mammary organoids subjected to replication stress activated a transcriptional response that selectively expands Brca2mut/WT luminal cells lacking hormone receptor expression (HR-). While CyTOF analyses revealed comparable epithelial compositions among wildtype and Brca2mut/WT mammary glands, Brca2mut/WT HR- luminal cells exhibited greater organoid formation and preferentially survived and expanded under replication stress. ScRNA-seq analysis corroborated the expansion of HR- luminal cells which express elevated levels of Tetraspanin-8 (Tspan8) and Thrsp mRNA, and pathways implicated in replication stress survival including Type I interferon responses. Notably, CRISPR/Cas9-mediated deletion of Tspan8 or Thrsp prevented Brca2mut/WT HR- luminal cell expansion. Our findings indicate that Brca2mut/WT cells have an activate a transcriptional response after replication stress that preferentially favours outgrowth of HR- luminal cells through the expression of interferon-responsive and mammary alveolar genes.