42 results on '"Izadmehr S"'
Search Results
2. Androgen Signaling in Metastatic Bone Disease
- Author
-
Arumugam, D., Izadmehr, S., and Alice Levine
- Published
- 2020
3. Anti-apoptotic BCL-2 proteins govern cellular outcome following B-RAFV600E inhibition and can be targeted to reduce resistance
- Author
-
Serasinghe, M N, primary, Missert, D J, additional, Asciolla, J J, additional, Podgrabinska, S, additional, Wieder, S Y, additional, Izadmehr, S, additional, Belbin, G, additional, Skobe, M, additional, and Chipuk, J E, additional
- Published
- 2014
- Full Text
- View/download PDF
4. Sensitization to the mitochondrial pathway of apoptosis augments melanoma tumor cell responses to conventional chemotherapeutic regimens
- Author
-
Anvekar, R A, primary, Asciolla, J J, additional, Lopez-Rivera, E, additional, Floros, K V, additional, Izadmehr, S, additional, Elkholi, R, additional, Belbin, G, additional, Sikora, A G, additional, and Chipuk, J E, additional
- Published
- 2012
- Full Text
- View/download PDF
5. Anti-apoptotic BCL-2 proteins govern cellular outcome following B-RAFV600E inhibition and can be targeted to reduce resistance.
- Author
-
Serasinghe, M N, Missert, D J, Asciolla, J J, Podgrabinska, S, Wieder, S Y, Izadmehr, S, Belbin, G, Skobe, M, and Chipuk, J E
- Subjects
APOPTOSIS inhibition ,BCL-2 proteins ,DRUG resistance in cancer cells ,RAF genes ,BRAF genes ,CARCINOGENS ,CELLULAR signal transduction ,ONCOGENES ,THERAPEUTICS - Abstract
In theory, pharmacological inhibition of oncogenic signaling is an effective strategy to halt cellular proliferation, induce apoptosis and eliminate cancer cells. In practice, drugs (for example, PLX-4032) that inhibit oncogenes like B-RAFV600E provide relatively short-term success in patients, owing to a combination of incomplete cellular responses and the development of resistance. To define the relationship between PLX-4032-induced responses and resistance, we interrogated the contributions of anti-apoptotic BCL-2 proteins in determining the fate of B-RAFV600E-inhibited melanoma cells. Although PLX-4032 eliminated B-RAFV600E signaling leading to marked cell cycle arrest, only a fraction of cells eventually underwent apoptosis. These data proposed two hypotheses regarding B-RAFV600E inhibition: (1) only a few cells generate a pro-apoptotic signal, or (2) all the cells generate a pro-apoptotic signal but the majority silences this pathway to ensure survival. Indeed, the latter hypothesis is supported by our observations as the addition of ABT-737, an inhibitor to anti-apoptotic BCL-2 proteins, revealed massive apoptosis following PLX-4032 exposure. B-RAFV600E inhibition alone sensitized cells to the mitochondrial pathway of apoptosis characterized by the rapid accumulation of BIM on the outer mitochondrial membrane, which could be functionally revealed by ABT-737 to promote apoptosis and loss of clonogenic survival. Furthermore, PLX-4032-resistant cells demonstrated collateral resistance to conventional chemotherapy, yet could be re-sensitized to PLX-4032 by BCL-2 family inhibition in vivo and conventional chemotherapies in vitro. Our data suggest that inhibiting anti-apoptotic BCL-2 proteins will enhance primary responses to PLX-4032, along with reducing the development of resistance to both targeted and conventional therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
6. HLA-E and NKG2A Mediate Resistance to M. bovis BCG Immunotherapy in Non-Muscle-Invasive Bladder Cancer.
- Author
-
Ranti D, Yu H, Wang YA, Bieber C, Strandgaard T, Salomé B, Houghton S, Kim J, Ravichandran H, Okulate I, Merritt E, Bang S, Demetriou A, Li Z, Lindskrog SV, Ruan DF, Daza J, Rai R, Hegewisch-Solloa E, Mace EM, Fernandez-Rodriguez R, Izadmehr S, Doherty G, Narasimhan A, Farkas AM, Cruz-Encarnacion P, Shroff S, Patel F, Tran M, Park SJ, Qi J, Patel M, Geanon D, Kelly G, de Real RM, Lee B, Nie K, Miake-Iye S, Angeliadis K, Radkevich E, Thin TH, Garcia-Barros M, Brown H, Martin B, Mateo A, Soto A, Sussman R, Shiwlani S, Francisco-Simon S, Beaumont KG, Hu Y, Wang YC, Wang L, Sebra RP, Smith S, Skobe M, Clancy-Thompson E, Palmer D, Hammond S, Hopkins BD, Wiklund P, Zhu J, Bravo-Cordero JJ, Brody R, Hopkins B, Chen Z, Kim-Schulze S, Dyrskjøt L, Elemento O, Tocheva A, Song WM, Bhardwaj N, Galsky MD, Sfakianos JP, and Horowitz A
- Abstract
Mycobacterium bovis Bacillus Calmette-Guerin (BCG) is the primary treatment for non-muscle-invasive bladder cancer (NMIBC), known to stimulate inflammatory cytokines, notably interferon (IFN)-γ. We observed that prolonged IFN-γ exposure fosters adaptive resistance in recurrent tumors, aiding immune evasion and tumor proliferation. We identify HLA-E and NKG2A, part of a novel NK and T cell checkpoint pathway, as key mediators of resistance in BCG-unresponsive NMIBC. IFN-γ enhances HLA-E and PD-L1 expression in recurrent tumors, with an enrichment of intra-tumoral NKG2A-expressing NK and CD8 T cells. CXCL9
+ macrophages and dendritic cells and CXCL12-expressing stromal cells likely recruit CXCR3/CXCR4-expressing NK and T cells and CXCR7+ HLA-EHIGH tumor cells. NK and CD8 T cells remain functional within BCG-unresponsive tumors but are inhibited by HLA-E and PD-L1, providing a framework for combined NKG2A and PD-L1 blockade strategy for bladder-sparing treatment of BCG-unresponsive NMIBC., Competing Interests: Conflict of Interest Disclosure Statement: The authors declare no potential conflicts of interest.- Published
- 2024
- Full Text
- View/download PDF
7. Single-cell transcriptomic-informed deconvolution of bulk data identifies immune checkpoint blockade resistance in urothelial cancer.
- Author
-
Wang L, Izadmehr S, Sfakianos JP, Tran M, Beaumont KG, Brody R, Cordon-Cardo C, Horowitz A, Sebra R, Oh WK, Bhardwaj N, Galsky MD, and Zhu J
- Abstract
Interactions within the tumor microenvironment (TME) significantly influence tumor progression and treatment responses. While single-cell RNA sequencing (scRNA-seq) and spatial genomics facilitate TME exploration, many clinical cohorts are assessed at the bulk tissue level. Integrating scRNA-seq and bulk tissue RNA-seq data through computational deconvolution is essential for obtaining clinically relevant insights. Our method, ProM, enables the examination of major and minor cell types. Through evaluation against existing methods using paired single-cell and bulk RNA sequencing of human urothelial cancer (UC) samples, ProM demonstrates superiority. Application to UC cohorts treated with immune checkpoint inhibitors reveals pre-treatment cellular features associated with poor outcomes, such as elevated SPP1 expression in macrophage/monocytes (MM). Our deconvolution method and paired single-cell and bulk tissue RNA-seq dataset contribute novel insights into TME heterogeneity and resistance to immune checkpoint blockade., Competing Interests: R.S. is a paid consultant and shareholder of GeneDx, Stamford, CT. A.H. receives research funds from Zumutor Biologics and is on the advisory boards of HTG Molecular Diagnostics, Immunorizon, UroGen, and Takeda. N.B. is an extramural member of the Parker Institute for Cancer Immunotherapy; receives research funds from Regeneron, Harbor Biomedical, DC Prime, and Dragonfly Therapeutics; and is on the advisory boards of Neon Therapeutics, Novartis, Avidea, Boehringer Ingelheim, Rome Therapeutics, Rubius Therapeutics, Roswell Park Comprehensive Cancer Center, BreakBio, Carisma Therapeutics, CureVac, Genotwin, BioNTech, Gilead Therapeutics, Tempest Therapeutics, and the Cancer Research Institute. M.D.G. has received grants or contracts from Bristol Myers Squibb, Novartis, Dendreon, AstraZeneca, and Merck; and has received consulting fees from Bristol Myers Squibb, Merck, Genentech, Inc., AstraZeneca, Pfizer, EMD Serono, Seagen, Janssen, Numab, Dragonfly, GlaxoSmithKline, Basilea, UroGen, RapptaTherapeutics, Alligator, Silverback, Fujifilm, and Curis., (© 2024 Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
8. C-reactive protein (CRP) as a prognostic biomarker in patients with urothelial carcinoma: A systematic review and meta-analysis.
- Author
-
Fujiwara Y, Karol AB, Joshi H, Reford E, Izadmehr S, Doroshow DB, and Galsky MD
- Subjects
- Humans, Carcinoma, Transitional Cell blood, Carcinoma, Transitional Cell pathology, Carcinoma, Transitional Cell diagnosis, Carcinoma, Transitional Cell mortality, Carcinoma, Transitional Cell drug therapy, Prognosis, Urinary Bladder Neoplasms mortality, Urinary Bladder Neoplasms blood, Urinary Bladder Neoplasms pathology, Urinary Bladder Neoplasms diagnosis, Urologic Neoplasms mortality, Urologic Neoplasms pathology, Urologic Neoplasms diagnosis, Urologic Neoplasms blood, Biomarkers, Tumor blood, C-Reactive Protein analysis
- Abstract
C-reactive protein (CRP) may reflect a pro-inflammatory tumor microenvironment and could represent a biomarker to select patients with urothelial carcinoma more likely to benefit from therapies directed at modulating tumor-promoting inflammation. We performed a systematic review to evaluate survival outcomes based on pre-treatment CRP values in urothelial carcinoma. The hazard ratios (HRs) of survival such as overall survival (OS) and progression-free survival (PFS) between groups with high versus low CRP values were pooled by the random-effect model meta-analyses. Overall, 28 studies comprising 6789 patients were identified for meta-analyses. High CRP levels were associated with shorter OS (HR=1.96 [95% CI: 1.64-2.33], p < 0.01), particularly in advanced disease treated with immune checkpoint blockade (ICB, HR=1.78 [1.47-2.15], p < 0.01). Similar findings were observed in ICB-treated patients with PFS. These findings suggest that CRP could be an attractive biomarker to select patients with urothelial carcinoma for strategies seeking to modulate tumor-promoting inflammation., Competing Interests: Declaration of Competing Interest None of the authors have a conflict of interest to report for the submitted work. D.B.D. reports a consulting advisory role for: Mirati, AstraZeneca, Summit Therapeutics, G1 Therapeutics, Sonata Therapeutics, Sanofi. M.D.G. reports stock from Rappta Therapeutics; a consulting/advisory role for BioMotiv, Janssen, Dendreon, Merck, GlaxoSmithKline, Lilly, Astellas Pharma, Genentech, Bristol-Myers Squibb, Novartis, Pfizer, EMD Serono, AstraZeneca, Seattle Genetics, Incyte, Aileron Therapeutics, Dracen, Inovio Pharmaceuticals, NuMab, Dragonfly Therapeutics, Basilea, Urogen, Infinity Pharmaceuticals, and Gilead; and institutional research funding from Janssen Oncology, Dendreon, Novartis, Bristol-Myers Squibb, Merck, AstraZeneca, and Genentech/Roche., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
9. Author Correction: Gemcitabine and cisplatin plus nivolumab as organ-sparing treatment for muscle-invasive bladder cancer: a phase 2 trial.
- Author
-
Galsky MD, Daneshmand S, Izadmehr S, Gonzalez-Kozlova E, Chan KG, Lewis S, Achkar BE, Dorff TB, Cetnar JP, Neil BO, D'Souza A, Mamtani R, Kyriakopoulos C, Jun T, Gogerly-Moragoda M, Brody R, Xie H, Nie K, Kelly G, Horowitz A, Kinoshita Y, Ellis E, Nose Y, Ioannou G, Cabal R, Del Valle DM, Haines GK, Wang L, Mouw KW, Samstein RM, Mehrazin R, Bhardwaj N, Yu M, Zhao Q, Kim-Schulze S, Sebra R, Zhu J, Gnjatic S, Sfakianos J, and Pal SK
- Published
- 2024
- Full Text
- View/download PDF
10. ACE2 and TMPRSS2 distribution in the respiratory tract of different animal species and its correlation with SARS-CoV-2 tissue tropism.
- Author
-
Carossino M, Izadmehr S, Trujillo JD, Gaudreault NN, Dittmar W, Morozov I, Balasuriya UBR, Cordon-Cardo C, García-Sastre A, and Richt JA
- Subjects
- Humans, Animals, SARS-CoV-2, Angiotensin-Converting Enzyme 2, Respiratory System, RNA, Messenger, Tropism, Serine Endopeptidases, COVID-19, Deer
- Abstract
A wide range of animal species show variable susceptibility to SARS-CoV-2; however, host factors associated with varied susceptibility remain to be defined. Here, we examined whether susceptibility to SARS-CoV-2 and virus tropism in different animal species are dependent on the expression and distribution of the virus receptor angiotensin-converting enzyme 2 ( ACE2 ) and the host cell factor transmembrane serine protease 2 ( TMPRSS2 ). We cataloged the upper and lower respiratory tract of multiple animal species and humans in a tissue-specific manner and quantitatively evaluated the distribution and abundance of ACE2 and TMPRSS2 mRNA in situ . Our results show that: (i) ACE2 and TMPRSS2 mRNA are abundant in the conduction portion of the respiratory tract, (ii) ACE2 mRNA occurs at a lower abundance compared to TMPRSS2 mRNA, (iii) co-expression of ACE2-TMPRSS2 mRNAs is highest in those species with the highest susceptibility to SARS-CoV-2 infection (i.e., cats, Syrian hamsters, and white-tailed deer), and (iv) expression of ACE2 and TMPRSS2 mRNA was not altered following SARS-CoV-2 infection. Our results demonstrate that while specific regions of the respiratory tract are enriched in ACE2 and TMPRSS2 mRNAs in different animal species, this is only a partial determinant of susceptibility to SARS-CoV-2 infection.IMPORTANCESARS-CoV-2 infects a wide array of domestic and wild animals, raising concerns regarding its evolutionary dynamics in animals and potential for spillback transmission of emerging variants to humans. Hence, SARS-CoV-2 infection in animals has significant public health relevance. Host factors determining animal susceptibility to SARS-CoV-2 are vastly unknown, and their characterization is critical to further understand susceptibility and viral dynamics in animal populations and anticipate potential spillback transmission. Here, we quantitatively assessed the distribution and abundance of the two most important host factors, angiotensin-converting enzyme 2 and transmembrane serine protease 2, in the respiratory tract of various animal species and humans. Our results demonstrate that while specific regions of the respiratory tract are enriched in these two host factors, they are only partial determinants of susceptibility. Detailed analysis of additional host factors is critical for our understanding of the underlying mechanisms governing viral susceptibility and reservoir hosts., Competing Interests: The J.A.R. laboratory received support from Tonix Pharmaceuticals, Xing Technologies, and Zoetis, outside of the reported work. J.A.R. developed patents and patent applications on the use of antivirals and vaccines for the treatment and prevention of virus infections, owned by Kansas State University. The other authors have declared that no conflict of interest exists.
- Published
- 2024
- Full Text
- View/download PDF
11. Cooperativity of c-MYC with Krüppel-Like Factor 6 Splice Variant 1 induces phenotypic plasticity and promotes prostate cancer progression and metastasis.
- Author
-
Izadmehr S, Fernandez-Hernandez H, Wiredja D, Kirschenbaum A, Lee-Poturalski C, Tavassoli P, Yao S, Schlatzer D, Hoon D, Difeo A, Levine AC, Mosquera JM, Galsky MD, Cordon-Cardo C, and Narla G
- Abstract
Metastasis remains a major cause of morbidity and mortality in men with prostate cancer, and the functional impact of the genetic alterations, alone or in combination, driving metastatic disease remains incompletely understood. The proto-oncogene c-MYC, commonly deregulated in prostate cancer. Transgenic expression of c-MYC is sufficient to drive the progression to prostatic intraepithelial neoplasia and ultimately to moderately differentiated localized primary tumors, however, c-MYC-driven tumors are unable to progress through the metastatic cascade, suggesting that a "second-hit" is necessary in the milieu of aberrant c-MYC-driven signaling. Here, we identified cooperativity between c-MYC and KLF6-SV1, an oncogenic splice variant of the KLF6 gene. Transgenic mice that co-expressed KLF6-SV1 and c-MYC developed progressive and metastatic prostate cancer with a histological and molecular phenotype like human prostate cancer. Silencing c-MYC expression significantly reduced tumor burden in these mice supporting the necessity for c-MYC in tumor maintenance. Unbiased global proteomic analysis of tumors from these mice revealed significantly enriched vimentin, a dedifferentiation and pro-metastatic marker, induced by KLF6-SV1. c-MYC-positive tumors were also significantly enriched for KLF6-SV1 in human prostate cancer specimens. Our findings provide evidence that KLF6-SV1 is an enhancer of c-MYC-driven prostate cancer progression and metastasis, and a correlated genetic event in human prostate cancer with potential translational significance., Competing Interests: Conflict of Interest G.N. is an author on patent 20090325150 (KLF6 alternative splice forms and a germline KLF6 DNA Polymorphism associated with increased cancer risk) related to this work.
- Published
- 2024
- Full Text
- View/download PDF
12. Gemcitabine and cisplatin plus nivolumab as organ-sparing treatment for muscle-invasive bladder cancer: a phase 2 trial.
- Author
-
Galsky MD, Daneshmand S, Izadmehr S, Gonzalez-Kozlova E, Chan KG, Lewis S, Achkar BE, Dorff TB, Cetnar JP, Neil BO, D'Souza A, Mamtani R, Kyriakopoulos C, Jun T, Gogerly-Moragoda M, Brody R, Xie H, Nie K, Kelly G, Horowitz A, Kinoshita Y, Ellis E, Nose Y, Ioannou G, Cabal R, Del Valle DM, Haines GK, Wang L, Mouw KW, Samstein RM, Mehrazin R, Bhardwaj N, Yu M, Zhao Q, Kim-Schulze S, Sebra R, Zhu J, Gnjatic S, Sfakianos J, and Pal SK
- Subjects
- Humans, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Deoxycytidine therapeutic use, Disease-Free Survival, Gemcitabine, Muscles, Neoadjuvant Therapy, Neoplasm Invasiveness, Nivolumab therapeutic use, Xeroderma Pigmentosum Group D Protein, Cisplatin therapeutic use, Urinary Bladder Neoplasms drug therapy, Urinary Bladder Neoplasms pathology
- Abstract
Cystectomy is a standard treatment for muscle-invasive bladder cancer (MIBC), but it is life-altering. We initiated a phase 2 study in which patients with MIBC received four cycles of gemcitabine, cisplatin, plus nivolumab followed by clinical restaging. Patients achieving a clinical complete response (cCR) could proceed without cystectomy. The co-primary objectives were to assess the cCR rate and the positive predictive value of cCR for a composite outcome: 2-year metastasis-free survival in patients forgoing immediate cystectomy or
- Published
- 2023
- Full Text
- View/download PDF
13. COVID-19 Therapeutics: Use, Mechanism of Action, and Toxicity (Vaccines, Monoclonal Antibodies, and Immunotherapeutics).
- Author
-
Chary M, Barbuto AF, Izadmehr S, Tarsillo M, Fleischer E, and Burns MM
- Subjects
- Female, Humans, Male, Pandemics prevention & control, SARS-CoV-2, Antibodies, Monoclonal therapeutic use, COVID-19 therapy, COVID-19 Serotherapy, Immunotherapy, Vaccines
- Abstract
SARS-CoV-2 emerged in December 2019 and led to the COVID-19 pandemic. Efforts to develop therapeutics have led to innovations such as mRNA vaccines and oral antivirals. Here we provide a narrative review of the biologic therapeutics used or proposed to treat COVID-19 during the last 3 years. This paper, along with its companion that covers xenobiotics and alternative remedies, is an update to our 2020 paper. Monoclonal antibodies prevent progression to severe disease, are not equally effective across variants, and are associated with minimal and self-limited reactions. Convalescent plasma has side effects like monoclonal antibodies, but with more infusion reactions and less efficacy. Vaccines prevent progression for a larger part of the population. DNA and mRNA vaccines are more effective than protein or inactivated virus vaccines. After mRNA vaccines, young men are more likely to have myocarditis in the subsequent 7 days. After DNA vaccines, those aged 30-50 are very slightly more likely to have thrombotic disease. To all vaccines we discuss, women are slightly more likely to have an anaphylactic reaction than men, but the absolute risk is small., (© 2023. American College of Medical Toxicology.)
- Published
- 2023
- Full Text
- View/download PDF
14. COVID-19 Therapeutics: Use, Mechanism of Action, and Toxicity (Xenobiotics).
- Author
-
Chary MA, Barbuto AF, Izadmehr S, Tarsillo M, Fleischer E, and Burns MM
- Subjects
- Humans, SARS-CoV-2, Xenobiotics, Pandemics prevention & control, Ivermectin therapeutic use, Antiviral Agents therapeutic use, COVID-19
- Abstract
SARS-CoV-2 emerged in 2019 and led to the COVID-19 pandemic. Efforts to develop therapeutics against SARS-Cov-2 led to both new treatments and attempts to repurpose existing medications. Here, we provide a narrative review of the xenobiotics and alternative remedies used or proposed to treat COVID-19. Most repositioned xenobiotics have had neither the feared toxicity nor the anticipated efficacy. Repurposed viral replication inhibitors are not efficacious and frequently associated with nausea, vomiting, and diarrhea. Antiviral medications designed specifically against SARS-CoV-2 may prevent progression to severe disease in at-risk individuals and appear to have a wide therapeutic index. Colloidal silver, zinc, and ivermectin have no demonstrated efficacy. Ivermectin has a wide therapeutic index but is not efficacious and acquiring it from veterinary sources poses additional danger. Chloroquine has a narrow therapeutic index and no efficacy. A companion review covers vaccines, monoclonal antibodies, and immunotherapies. Together, these two reviews form an update to our 2020 review., (© 2022. American College of Medical Toxicology.)
- Published
- 2023
- Full Text
- View/download PDF
15. NKG2A and HLA-E define an alternative immune checkpoint axis in bladder cancer.
- Author
-
Salomé B, Sfakianos JP, Ranti D, Daza J, Bieber C, Charap A, Hammer C, Banchereau R, Farkas AM, Ruan DF, Izadmehr S, Geanon D, Kelly G, de Real RM, Lee B, Beaumont KG, Shroff S, Wang YA, Wang YC, Thin TH, Garcia-Barros M, Hegewisch-Solloa E, Mace EM, Wang L, O'Donnell T, Chowell D, Fernandez-Rodriguez R, Skobe M, Taylor N, Kim-Schulze S, Sebra RP, Palmer D, Clancy-Thompson E, Hammond S, Kamphorst AO, Malmberg KJ, Marcenaro E, Romero P, Brody R, Viard M, Yuki Y, Martin M, Carrington M, Mehrazin R, Wiklund P, Mellman I, Mariathasan S, Zhu J, Galsky MD, Bhardwaj N, and Horowitz A
- Subjects
- B7-H1 Antigen metabolism, CD8-Positive T-Lymphocytes, Histocompatibility Antigens Class I, Humans, Programmed Cell Death 1 Receptor, HLA-E Antigens, NK Cell Lectin-Like Receptor Subfamily C metabolism, Urinary Bladder Neoplasms therapy
- Abstract
Programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1)-blockade immunotherapies have limited efficacy in the treatment of bladder cancer. Here, we show that NKG2A associates with improved survival and responsiveness to PD-L1 blockade immunotherapy in bladder tumors that have high abundance of CD8
+ T cells. In bladder tumors, NKG2A is acquired on CD8+ T cells later than PD-1 as well as other well-established immune checkpoints. NKG2A+ PD-1+ CD8+ T cells diverge from classically defined exhausted T cells through their ability to react to human leukocyte antigen (HLA) class I-deficient tumors using T cell receptor (TCR)-independent innate-like mechanisms. HLA-ABC expression by bladder tumors is progressively diminished as disease progresses, framing the importance of targeting TCR-independent anti-tumor functions. Notably, NKG2A+ CD8+ T cells are inhibited when HLA-E is expressed by tumors and partly restored upon NKG2A blockade in an HLA-E-dependent manner. Overall, our study provides a framework for subsequent clinical trials combining NKG2A blockade with other T cell-targeted immunotherapies, where tumors express higher levels of HLA-E., Competing Interests: Declaration of interests L.W., R.P.S., and J.Z. are employees of Sema4. A.H. receives research funds from Zumutor Biologics and is on the advisory boards of HTG Molecular Diagnostics, Immunorizon, UroGen, and Takeda. N.B. is an extramural member of the Parker Institute for Cancer Immunotherapy; receives research funds from Regeneron, Harbor Biomedical, DC Prime, and Dragonfly Therapeutics; and is on the advisory boards of Neon Therapeutics, Novartis, Avidea, Boehringer Ingelheim, Rome Therapeutics, Rubius Therapeutics, Roswell Park Comprehensive Cancer Center, BreakBio, Carisma Therapeutics, CureVac, Genotwin, BioNTech, Gilead Therapeutics, Tempest Therapeutics, and the Cancer Research Institute. A patent related to this work was filed to the United States Patent and Trademark Office (63/313,823)., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
16. The Evolving Clinical Management of Genitourinary Cancers Amid the COVID-19 Pandemic.
- Author
-
Izadmehr S, Lundon DJ, Mohamed N, Katims A, Patel V, Eilender B, Mehrazin R, Badani KK, Sfakianos JP, Tsao CK, Wiklund P, Oh WK, Cordon-Cardo C, Tewari AK, Galsky MD, and Kyprianou N
- Abstract
Coronavirus disease-2019 (COVID-19), a disease caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection, has become an unprecedented global health emergency, with fatal outcomes among adults of all ages throughout the world. There is a high incidence of infection and mortality among cancer patients with evidence to support that patients diagnosed with cancer and SARS-CoV-2 have an increased likelihood of a poor outcome. Clinically relevant changes imposed as a result of the pandemic, are either primary, due to changes in timing or therapeutic modality; or secondary, due to altered cooperative effects on disease progression or therapeutic outcomes. However, studies on the clinical management of patients with genitourinary cancers during the COVID-19 pandemic are limited and do little to differentiate primary or secondary impacts of COVID-19. Here, we provide a review of the epidemiology and biological consequences of SARS-CoV-2 infection in GU cancer patients as well as the impact of COVID-19 on the diagnosis and management of these patients, and the use and development of novel and innovative diagnostic tests, therapies, and technology. This article also discusses the biomedical advances to control the virus and evolving challenges in the management of prostate, bladder, kidney, testicular, and penile cancers at all stages of the patient journey during the first year of the COVID-19 pandemic., Competing Interests: C-KT has financial relationships in consulting with Clovis, Pfizer, and Eisai. WO has financial relationships in consulting with Astellas, Astra Zeneca, Bayer, Janssen, Sanofi, Sema4, and TeneoBio. AT has financial relationships in consulting with Intuitive Surgical, Promaxo, Roivant, Siemens, and Kite Pharma. He serves as an advisor for and owns equity in the form of stock certificates in Promaxo. MG has financial relationships in consulting with BioMotiv, Janssen, Dendreon, Merck, GlaxoSmithKline, Lilly, Astellas Pharma, Genentech, Bristol‐Meyers Squibb, Novartis, Pfizer, EMD Serono, AstraZeneca, Seattle Genetics, Incyte, Aileron Therapeutics, Dracen, Inovio Pharmaceuticals, NuMab, and Dragonfly Therapeutics. Matthew Galsky has received research funding from Janssen Oncology, Dendreon, Novartis, Bristol‐Myers Squibb, Merck, AstraZeneca, and Genentech/Roche. MG also has financial ownership interest in Rappta Therapeutics. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Izadmehr, Lundon, Mohamed, Katims, Patel, Eilender, Mehrazin, Badani, Sfakianos, Tsao, Wiklund, Oh, Cordon-Cardo, Tewari, Galsky and Kyprianou.)
- Published
- 2021
- Full Text
- View/download PDF
17. Myeloid Cell-associated Resistance to PD-1/PD-L1 Blockade in Urothelial Cancer Revealed Through Bulk and Single-cell RNA Sequencing.
- Author
-
Wang L, Sfakianos JP, Beaumont KG, Akturk G, Horowitz A, Sebra RP, Farkas AM, Gnjatic S, Hake A, Izadmehr S, Wiklund P, Oh WK, Szabo PM, Wind-Rotolo M, Unsal-Kacmaz K, Yao X, Schadt E, Sharma P, Bhardwaj N, Zhu J, and Galsky MD
- Subjects
- Drug Resistance, Neoplasm, Humans, Carcinoma, Transitional Cell drug therapy, Carcinoma, Transitional Cell genetics, Immune Checkpoint Inhibitors therapeutic use, Myeloid Cells physiology, Sequence Analysis, RNA, Urinary Bladder Neoplasms drug therapy, Urinary Bladder Neoplasms genetics
- Abstract
Purpose: To define dominant molecular and cellular features associated with PD-1/PD-L1 blockade resistance in metastatic urothelial cancer., Experimental Design: We pursued an unbiased approach using bulk RNA sequencing data from two clinical trials to discover (IMvigor 210) and validate (CheckMate 275) pretreatment molecular features associated with resistance to PD-1/PD-L1 blockade in metastatic urothelial cancer. We then generated single-cell RNA sequencing (scRNA-seq) data from muscle-invasive bladder cancer specimens to dissect the cellular composition underlying the identified gene signatures., Results: We identified an adaptive immune response gene signature associated with response and a protumorigenic inflammation gene signature associated with resistance to PD-1/PD-L1 blockade. The adaptive immune response:protumorigenic inflammation signature expression ratio, coined the 2IR score, best correlated with clinical outcomes, and was externally validated. Mapping these bulk gene signatures onto scRNA-seq data uncovered their underlying cellular diversity, with prominent expression of the protumorigenic inflammation signature by myeloid phagocytic cells. However, heterogeneity in expression of adaptive immune and protumorigenic inflammation genes was observed among single myeloid phagocytic cells, quantified as the myeloid single cell immune:protumorigenic inflammation ratio (M
sc 2IR) score. Single myeloid phagocytic cells with low Msc 2IR scores demonstrated upregulation of proinflammatory cytokines/chemokines and downregulation of antigen presentation genes, were unrelated to M1 versus M2 polarization, and were enriched in pretreatment blood samples from patients with PD-L1 blockade-resistant metastatic urothelial cancer., Conclusions: The balance of adaptive immunity and protumorigenic inflammation in individual tumor microenvironments is associated with PD-1/PD-L1 resistance in urothelial cancer with the latter linked to a proinflammatory cellular state of myeloid phagocytic cells detectable in tumor and blood. See related commentary by Drake, p. 4139 ., (©2021 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
18. The SRG rat, a Sprague-Dawley Rag2/Il2rg double-knockout validated for human tumor oncology studies.
- Author
-
Noto FK, Sangodkar J, Adedeji BT, Moody S, McClain CB, Tong M, Ostertag E, Crawford J, Gao X, Hurst L, O'Connor CM, Hanson EN, Izadmehr S, Tohmé R, Narla J, LeSueur K, Bhattacharya K, Rupani A, Tayeh MK, Innis JW, Galsky MD, Evers BM, DiFeo A, Narla G, and Jamling TY
- Subjects
- Animals, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung pathology, Cell Line, Tumor, Gene Deletion, Humans, Lung Neoplasms genetics, Lung Neoplasms pathology, Neoplasms, Experimental genetics, Rats, Rats, Sprague-Dawley, Xenograft Model Antitumor Assays standards, Interleukin Receptor Common gamma Subunit genetics, Neoplasms, Experimental pathology, Xenograft Model Antitumor Assays methods
- Abstract
We have created the immunodeficient SRG rat, a Sprague-Dawley Rag2/Il2rg double knockout that lacks mature B cells, T cells, and circulating NK cells. This model has been tested and validated for use in oncology (SRG OncoRat®). The SRG rat demonstrates efficient tumor take rates and growth kinetics with different human cancer cell lines and PDXs. Although multiple immunodeficient rodent strains are available, some important human cancer cell lines exhibit poor tumor growth and high variability in those models. The VCaP prostate cancer model is one such cell line that engrafts unreliably and grows irregularly in existing models but displays over 90% engraftment rate in the SRG rat with uniform growth kinetics. Since rats can support much larger tumors than mice, the SRG rat is an attractive host for PDX establishment. Surgically resected NSCLC tissue from nine patients were implanted in SRG rats, seven of which engrafted and grew for an overall success rate of 78%. These developed into a large tumor volume, over 20,000 mm3 in the first passage, which would provide an ample source of tissue for characterization and/or subsequent passage into NSG mice for drug efficacy studies. Molecular characterization and histological analyses were performed for three PDX lines and showed high concordance between passages 1, 2 and 3 (P1, P2, P3), and the original patient sample. Our data suggest the SRG OncoRat is a valuable tool for establishing PDX banks and thus serves as an alternative to current PDX mouse models hindered by low engraftment rates, slow tumor growth kinetics, and multiple passages to develop adequate tissue banks., Competing Interests: Dr. Goutham Narla is on the scientific advisory board for HERA Biolabs. This does not alter our adherence to PLOS ONE policies on sharing data and materials.
- Published
- 2020
- Full Text
- View/download PDF
19. COVID-19: Therapeutics and Their Toxicities.
- Author
-
Chary MA, Barbuto AF, Izadmehr S, Hayes BD, and Burns MM
- Subjects
- Betacoronavirus drug effects, COVID-19, Humans, Pandemics, SARS-CoV-2, United States, Antiviral Agents therapeutic use, Antiviral Agents toxicity, Chloroquine therapeutic use, Chloroquine toxicity, Coronavirus Infections drug therapy, Hydroxychloroquine therapeutic use, Hydroxychloroquine toxicity, Pneumonia, Viral drug therapy
- Abstract
SARS-CoV-2 is a novel coronavirus that emerged in 2019 and is causing the COVID-19 pandemic. There is no current standard of care. Clinicians need to be mindful of the toxicity of a wide variety of possibly unfamiliar substances being tested or repurposed to treat COVID-19. The United States Food and Drug Administration (FDA) has provided emergency authorization for the use of chloroquine and hydroxychloroquine. These two medications may precipitate ventricular dysrhythmias, necessitating cardiac and electrolyte monitoring, and in severe cases, treatment with epinephrine and high-doses of diazepam. Recombinant protein therapeutics may cause serum sickness or immune complex deposition. Nucleic acid vaccines may introduce mutations into the human genome. ACE inhibitors and ibuprofen have been suggested to exacerbate the pathogenesis of COVID-19. Here, we review the use, mechanism of action, and toxicity of proposed COVID-19 therapeutics.
- Published
- 2020
- Full Text
- View/download PDF
20. Selective PP2A Enhancement through Biased Heterotrimer Stabilization.
- Author
-
Leonard D, Huang W, Izadmehr S, O'Connor CM, Wiredja DD, Wang Z, Zaware N, Chen Y, Schlatzer DM, Kiselar J, Vasireddi N, Schüchner S, Perl AL, Galsky MD, Xu W, Brautigan DL, Ogris E, Taylor DJ, and Narla G
- Subjects
- Amino Acid Sequence, Animals, Cell Line, Tumor, Enzyme Activators metabolism, HEK293 Cells, Heterografts, Humans, Male, Mice, Mice, Nude, Models, Molecular, Multiprotein Complexes metabolism, Protein Phosphatase 2 chemistry, Protein Subunits, Protein Phosphatase 2 metabolism
- Abstract
Impairment of protein phosphatases, including the family of serine/threonine phosphatases designated PP2A, is essential for the pathogenesis of many diseases, including cancer. The ability of PP2A to dephosphorylate hundreds of proteins is regulated by over 40 specificity-determining regulatory "B" subunits that compete for assembly and activation of heterogeneous PP2A heterotrimers. Here, we reveal how a small molecule, DT-061, specifically stabilizes the B56α-PP2A holoenzyme in a fully assembled, active state to dephosphorylate selective substrates, such as its well-known oncogenic target, c-Myc. Our 3.6 Å structure identifies molecular interactions between DT-061 and all three PP2A subunits that prevent dissociation of the active enzyme and highlight inherent mechanisms of PP2A complex assembly. Thus, our findings provide fundamental insights into PP2A complex assembly and regulation, identify a unique interfacial stabilizing mode of action for therapeutic targeting, and aid in the development of phosphatase-based therapeutics tailored against disease specific phospho-protein targets., Competing Interests: Declaration of Interests The Icahn School of Medicine at Mount Sinai has filed patents covering composition of matter on the small molecules disclosed herein for the treatment of human cancer and other diseases (International Application numbers PCT/US15/19770 and PCT/US15/19764 and US Patent number US 9,540,358 B2). Mount Sinai is actively seeking commercial partners for the further development of the technology. G.N. has a financial interest in the commercialization of the technology. RAPPTA Therapeutics has licensed the cryo-EM coordinates for the clinical and commercial development of novel series of small molecule PP2A activators from the University of Michigan (G.N.) and Case Western Reserve University (D.T.). G.N. and D.T. have an ownership interest in RAPPTA Therapeutics. D.L.B. functions as a SAB member for RAPPTA. The Medical University of Vienna, on behalf of E.O., is filing a patent on the PP2A methyl-C subunit specific monoclonal antibody 7C10 disclosed herein for the diagnostic use of 7C10., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
21. Protein phosphatase 2A activation as a therapeutic strategy for managing MYC-driven cancers.
- Author
-
Farrington CC, Yuan E, Mazhar S, Izadmehr S, Hurst L, Allen-Petersen BL, Janghorban M, Chung E, Wolczanski G, Galsky M, Sears R, Sangodkar J, and Narla G
- Subjects
- Apoptosis drug effects, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung pathology, Cell Line, Tumor, Cell Proliferation drug effects, Gene Expression Regulation, Neoplastic drug effects, Humans, Proteolysis drug effects, Proto-Oncogene Mas, Proto-Oncogene Proteins c-myc antagonists & inhibitors, Proto-Oncogene Proteins c-myc chemistry, Small Molecule Libraries pharmacology, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms pathology, Protein Phosphatase 2 genetics, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins p21(ras) genetics, Tumor Suppressor Proteins genetics
- Abstract
The tumor suppressor protein phosphatase 2A (PP2A) is a serine/threonine phosphatase whose activity is inhibited in most human cancers. One of the best-characterized PP2A substrates is MYC proto-oncogene basic helix-loop-helix transcription factor (MYC), whose overexpression is commonly associated with aggressive forms of this disease. PP2A directly dephosphorylates MYC, resulting in its degradation. To explore the therapeutic potential of direct PP2A activation in a diverse set of MYC-driven cancers, here we used biochemical assays, recombinant cell lines, gene expression analyses, and immunohistochemistry to evaluate a series of first-in-class small-molecule activators of PP2A (SMAPs) in Burkitt lymphoma, KRAS-driven non-small cell lung cancer, and triple-negative breast cancer. In all tested models of MYC-driven cancer, the SMAP treatment rapidly and persistently inhibited MYC expression through proteasome-mediated degradation, inhibition of MYC transcriptional activity, decreased cancer cell proliferation, and tumor growth inhibition. Importantly, we generated a series of cell lines expressing PP2A-dependent phosphodegron variants of MYC and demonstrated that the antitumorigenic activity of SMAPs depends on MYC degradation. Collectively, the findings presented here indicate a pharmacologically tractable approach to drive MYC degradation by using SMAPs for the management of a broad range of MYC-driven cancers., (© 2020 Farrington et al.)
- Published
- 2020
- Full Text
- View/download PDF
22. MIFEPRISTONE TREATMENT FOR MILD AUTONOMOUS CORTISOL SECRETION DUE TO ADRENAL ADENOMAS: A PILOT STUDY.
- Author
-
Belokovskaya R, Ravikumar A, Arumugam D, Izadmehr S, Goddard GM, Geer EB, and Levine AC
- Subjects
- Humans, Hydrocortisone, Pilot Projects, Quality of Life, Adenoma drug therapy, Adrenal Gland Neoplasms drug therapy, Mifepristone therapeutic use
- Abstract
Objective: Adrenal incidentalomas are increasingly detected with the widespread use of thoracic and abdominal imaging. The most common secretory syndrome in adrenal nodules is autonomous cortisol secretion (ACS). Recent data show that even mild cortisol excess is associated with adverse outcomes. The glucocorticoid receptor antagonist mifepristone has been used in patients with overt Cushing syndrome and hyperglycemia. The purpose of our study was to determine the effect of mifepristone on metabolic parameters in patients with ACS and concomitant prediabetes or diabetes. Methods: Eight patients with either unilateral or bilateral adrenal nodules with ACS were included in the study. Fasting laboratory tests including glucose and insulin levels to calculate homeostatic model assessment for insulin resistance (HOMA-IR) were performed at baseline and again after either 3 months (3 patients) or 6 months (5 patients) on mifepristone 300 mg daily treatment. Patients also completed several validated surveys on mood and quality of life at baseline and follow-up. Results: There were significant reductions in fasting glucose measurements and insulin resistance as measured by HOMA-IR in the 6 of 8 study patients in whom these measurements were available ( P = .03). Conclusion: This pilot study demonstrates that mifepristone treatment of ACS is associated with a significant decrease in fasting glucose and insulin resistance as measured by HOMA-IR scores. Mifepristone treatment of ACS may be considered as a medical option for patients with ACS due to adrenal adenomas with concomitant abnormal glucose parameters in whom surgical removal is not being considered. Abbreviations: ACS = autonomous cortisol secretion; ACTH = adrenocorticotropic hormone; AI = adrenal incidentaloma; DHEAS = dehydroepiandrosterone sulfate; GR = glucocorticoid receptor; HbA1c = hemoglobin A1c; HOMA-IR = homeostatic model assessment for insulin resistance; ODT = overnight dexamethasone suppression test; QoL = quality of life; STAI = state trait anxiety inventory; TSH = thyroid stimulating hormone; UFC = urinary free cortisol.
- Published
- 2019
- Full Text
- View/download PDF
23. Isolation and Characterization of Tumor-initiating Cells from Sarcoma Patient-derived Xenografts.
- Author
-
Han D, Rodriguez-Bravo V, Izadmehr S, Domingo-Domenech J, and Cordon-Cardo C
- Subjects
- Animals, Cell Differentiation, Cell Line, Tumor, Disease Models, Animal, Heterografts, Humans, Mice, Neoplastic Stem Cells pathology, Sarcoma pathology
- Abstract
The existence and importance of tumor-initiating cells (TICs) have been supported by increasing evidence during the past decade. These TICs have been shown to be responsible for tumor initiation, metastasis, and drug resistance. Therefore, it is important to develop specific TIC-targeting therapy in addition to current chemotherapy strategies, which mostly focus on the bulk of non-TICs. In order to further understand the mechanism behind the malignancy of TICs, we describe a method to isolate and to characterize TICs in human sarcomas. Herein, we show a detailed protocol to generate patient-derived xenografts (PDXs) of human sarcomas and to isolate TICs by fluorescence-activated cell sorting (FACS) using human leukocyte antigen class I (HLA-1) as a negative marker. Also, we describe how to functionally characterize these TICs, including a sphere formation assay and a tumor formation assay, and to induce differentiation along mesenchymal pathways. The isolation and characterization of PDX TICs provide clues for the discovery of potential targeting therapy reagents. Moreover, increasing evidence suggests that this protocol may be further extended to isolate and characterize TICs from other types of human cancers.
- Published
- 2019
- Full Text
- View/download PDF
24. Direct activation of PP2A for the treatment of tyrosine kinase inhibitor-resistant lung adenocarcinoma.
- Author
-
Tohmé R, Izadmehr S, Gandhe S, Tabaro G, Vallabhaneni S, Thomas A, Vasireddi N, Dhawan NS, Ma'ayan A, Sharma N, Galsky MD, Ohlmeyer M, Sangodkar J, and Narla G
- Subjects
- Adenocarcinoma of Lung genetics, Adenocarcinoma of Lung pathology, Animals, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Apoptosis drug effects, Apoptosis genetics, Cell Line, Tumor, Cell Proliferation drug effects, Cell Proliferation genetics, Drug Resistance, Neoplasm genetics, Enzyme Activators therapeutic use, ErbB Receptors antagonists & inhibitors, ErbB Receptors genetics, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, Lung Neoplasms genetics, Lung Neoplasms pathology, MAP Kinase Signaling System drug effects, MAP Kinase Signaling System genetics, Male, Mice, Phosphatidylinositol 3-Kinase metabolism, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Xenograft Model Antitumor Assays, Adenocarcinoma of Lung drug therapy, Antineoplastic Combined Chemotherapy Protocols pharmacology, Drug Resistance, Neoplasm drug effects, Enzyme Activators pharmacology, Lung Neoplasms drug therapy, Phosphoprotein Phosphatases metabolism
- Abstract
Although tyrosine kinase inhibitors (TKIs) have demonstrated significant efficacy in advanced lung adenocarcinoma (LUAD) patients with pathogenic alterations in EGFR, most patients develop acquired resistance to these agents via mechanisms enabling the sustained activation of the PI3K and MAPK oncogenic pathways downstream of EGFR. The tumor suppressor protein phosphatase 2A (PP2A) acts as a negative regulator of these pathways. We hypothesize that activation of PP2A simultaneously inhibits the PI3K and MAPK pathways and represents a promising therapeutic strategy for the treatment of TKI-resistant LUAD. After establishing the efficacy of small molecule activators of PP2A (SMAPs) in a transgenic EGFRL858R model and TKI-sensitive cell lines, we evaluated their therapeutic potential in vitro and in vivo in TKI-resistant models. PP2A activation resulted in apoptosis, significant tumor growth inhibition, and downregulation of PI3K and MAPK pathways. Combination of SMAPs and TKI afatinib resulted in an enhanced effect on the downregulation of the PI3K pathway via degradation of the PP2A endogenous inhibitor CIP2A. An improved effect on tumor growth inhibition was observed in a TKI-resistant xenograft mouse model treated with a combination of both agents. These collective data support the development of PP2A activators for the treatment of TKI-resistant LUAD.
- Published
- 2019
- Full Text
- View/download PDF
25. Mechanisms of Osteoblastic Bone Metastasis in Prostate Cancer: Role of Prostatic Acid Phosphatase.
- Author
-
Quiroz-Munoz M, Izadmehr S, Arumugam D, Wong B, Kirschenbaum A, and Levine AC
- Abstract
Prostate cancer (PCa) preferentially metastasizes to bone, leading to complications including severe pain, fractures, spinal cord compression, bone marrow suppression, and a mortality of ∼70%. In spite of recent advances in chemo-, hormonal, and radiation therapies, bone-metastatic, castrate-resistant PCa is incurable. PCa is somewhat unique among the solid tumors in its tendency to produce osteoblastic lesions composed of hypermineralized bone with multiple layers of poorly organized type I collagen fibrils that have reduced mechanical strength. Many of the signaling pathways that control normal bone homeostasis are at play in pathologic PCa bone metastases, including the receptor activator of nuclear factor- κ B/receptor activator of nuclear factor- κ B ligand/osteoprotegerin system. A number of PCa-derived soluble factors have been shown to induce the dysfunctional osteoblastic phenotype. However, therapies directed at these osteoblastic-stimulating proteins have yielded disappointing clinical results to date. One of the soluble factors expressed by PCa cells, particularly in bone metastases, is prostatic acid phosphatase (PAP). Human PAP is a prostate epithelium-specific secretory protein that was the first tumor marker ever described. Biologically, PAP exhibits both phosphatase activity and ecto-5'-nucleotidase activity, generating extracellular phosphate and adenosine as the final products. Accumulating evidence indicates that PAP plays a causal role in the osteoblastic phenotype and aberrant bone mineralization seen in bone-metastatic, castrate-resistant PCa. Targeting PAP may represent a therapeutic approach to improve morbidity and mortality from PCa osteoblastic bone metastases.
- Published
- 2019
- Full Text
- View/download PDF
26. Sequential trafficking of Env and Gag to HIV-1 T cell virological synapses revealed by live imaging.
- Author
-
Wang L, Izadmehr S, Kamau E, Kong XP, and Chen BK
- Subjects
- CD4-Positive T-Lymphocytes physiology, Genes, Reporter, Green Fluorescent Proteins analysis, Green Fluorescent Proteins genetics, Humans, Jurkat Cells, Protein Transport, Recombinant Fusion Proteins analysis, Recombinant Fusion Proteins genetics, Staining and Labeling, Time-Lapse Imaging, CD4-Positive T-Lymphocytes virology, Cell Adhesion, HIV-1 physiology, Intravital Microscopy, Virus Assembly, env Gene Products, Human Immunodeficiency Virus metabolism, gag Gene Products, Human Immunodeficiency Virus metabolism
- Abstract
Background: HIV infection is enhanced by cell adhesions that form between infected and uninfected T cells called virological synapses (VS). VS are initiated by an interaction between Env and CD4 on cell surfaces and result in the recruitment of virus assembly to the site of cell-cell contact. However, the recruitment of Env to the VS and its relationship to Gag recruitment is not well defined., Results: To study the trafficking of HIV-1 Env through the VS, we constructed a molecular clone of HIV carrying a green fluorescent protein-Env fusion protein called, HIV Env-isfGFP-∆V1V2. The Env-isfGFP-∆V1V2 fusion protein does not produce virus particles on its own, but can be rescued by cotransfection with full-length HIV constructs and produce virus particles that package the fluorescent Env. These rescued fluorescent Env can participate in VS formation and can be used to directly image CD4-dependent Env transfer across VS from donor to target cells. The movements of fluorescently tagged Gag and Env to the VS and transfer into target cells can be also tracked through live imaging. Time lapse live imaging reveals evidence of limited Env accumulation at the site of cell-cell contact shortly after cell adhesion, followed by Gag re-distribution to contact area. Both Gag and Env can be recruited to form button-like spots characteristic of VS., Conclusions: Env and Gag are recruited to the VS in a coordinated temporal sequence and subsequently transfer together across the synapse into the target cell. Env accumulations, when observed, are earlier than Gag re-distribution to the contact area during formation of VS.
- Published
- 2019
- Full Text
- View/download PDF
27. PP2A inhibition is a druggable MEK inhibitor resistance mechanism in KRAS-mutant lung cancer cells.
- Author
-
Kauko O, O'Connor CM, Kulesskiy E, Sangodkar J, Aakula A, Izadmehr S, Yetukuri L, Yadav B, Padzik A, Laajala TD, Haapaniemi P, Momeny M, Varila T, Ohlmeyer M, Aittokallio T, Wennerberg K, Narla G, and Westermarck J
- Subjects
- Animals, Cell Line, Tumor, Cellular Senescence drug effects, Humans, Lung Neoplasms pathology, MAP Kinase Signaling System drug effects, Male, Mice, Inbred BALB C, Mice, Nude, Mitogen-Activated Protein Kinase Kinases metabolism, Protein Kinase Inhibitors pharmacology, Protein Phosphatase 2 metabolism, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction drug effects, TOR Serine-Threonine Kinases metabolism, Drug Resistance, Neoplasm drug effects, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Mutation genetics, Protein Kinase Inhibitors therapeutic use, Protein Phosphatase 2 antagonists & inhibitors, Proto-Oncogene Proteins p21(ras) genetics
- Abstract
Kinase inhibitor resistance constitutes a major unresolved clinical challenge in cancer. Furthermore, the role of serine/threonine phosphatase deregulation as a potential cause for resistance to kinase inhibitors has not been thoroughly addressed. We characterize protein phosphatase 2A (PP2A) activity as a global determinant of KRAS-mutant lung cancer cell resistance across a library of >200 kinase inhibitors. The results show that PP2A activity modulation alters cancer cell sensitivities to a large number of kinase inhibitors. Specifically, PP2A inhibition ablated mitogen-activated protein kinase kinase (MEK) inhibitor response through the collateral activation of AKT/mammalian target of rapamycin (mTOR) signaling. Combination of mTOR and MEK inhibitors induced cytotoxicity in PP2A-inhibited cells, but even this drug combination could not abrogate MYC up-regulation in PP2A-inhibited cells. Treatment with an orally bioavailable small-molecule activator of PP2A DT-061, in combination with the MEK inhibitor AZD6244, resulted in suppression of both p-AKT and MYC, as well as tumor regression in two KRAS-driven lung cancer mouse models. DT-061 therapy also abrogated MYC-driven tumorigenesis. These data demonstrate that PP2A deregulation drives MEK inhibitor resistance in KRAS-mutant cells. These results emphasize the need for better understanding of phosphatases as key modulators of cancer therapy responses., (Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2018
- Full Text
- View/download PDF
28. Small-Molecule Activators of Protein Phosphatase 2A for the Treatment of Castration-Resistant Prostate Cancer.
- Author
-
McClinch K, Avelar RA, Callejas D, Izadmehr S, Wiredja D, Perl A, Sangodkar J, Kastrinsky DB, Schlatzer D, Cooper M, Kiselar J, Stachnik A, Yao S, Hoon D, McQuaid D, Zaware N, Gong Y, Brautigan DL, Plymate SR, Sprenger CCT, Oh WK, Levine AC, Kirschenbaum A, Sfakianos JP, Sears R, DiFeo A, Ioannou Y, Ohlmeyer M, Narla G, and Galsky MD
- Subjects
- Animals, Cell Line, Tumor, Enzyme Activators pharmacology, Heterografts, Humans, Male, Mice, Mice, SCID, Phosphoproteins metabolism, Protein Phosphatase 2C metabolism, Proteomics, RNA, Messenger genetics, Receptors, Androgen genetics, Receptors, Androgen metabolism, Small Molecule Libraries pharmacology, Enzyme Activators therapeutic use, Prostatic Neoplasms, Castration-Resistant drug therapy, Prostatic Neoplasms, Castration-Resistant enzymology, Protein Phosphatase 2C drug effects, Small Molecule Libraries therapeutic use
- Abstract
Primary prostate cancer is generally treatable by androgen deprivation therapy, however, later recurrences of castrate-resistant prostate cancer (CRPC) that are more difficult to treat nearly always occur due to aberrant reactivation of the androgen receptor (AR). In this study, we report that CRPC cells are particularly sensitive to the growth-inhibitory effects of reengineered tricyclic sulfonamides, a class of molecules that activate the protein phosphatase PP2A, which inhibits multiple oncogenic signaling pathways. Treatment of CRPC cells with small-molecule activators of PP2A (SMAP) in vitro decreased cellular viability and clonogenicity and induced apoptosis. SMAP treatment also induced an array of significant changes in the phosphoproteome, including most notably dephosphorylation of full-length and truncated isoforms of the AR and downregulation of its regulatory kinases in a dose-dependent and time-dependent manner. In murine xenograft models of human CRPC, the potent compound SMAP-2 exhibited efficacy comparable with enzalutamide in inhibiting tumor formation. Overall, our results provide a preclinical proof of concept for the efficacy of SMAP in AR degradation and CRPC treatment. Significance: A novel class of small-molecule activators of the tumor suppressor PP2A, a serine/threonine phosphatase that inhibits many oncogenic signaling pathways, is shown to deregulate the phosphoproteome and to destabilize the androgen receptor in advanced prostate cancer. Cancer Res; 78(8); 2065-80. ©2018 AACR ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
29. Factor XII and uPAR upregulate neutrophil functions to influence wound healing.
- Author
-
Stavrou EX, Fang C, Bane KL, Long AT, Naudin C, Kucukal E, Gandhi A, Brett-Morris A, Mumaw MM, Izadmehr S, Merkulova A, Reynolds CC, Alhalabi O, Nayak L, Yu WM, Qu CK, Meyerson HJ, Dubyak GR, Gurkan UA, Nieman MT, Sen Gupta A, Renné T, and Schmaier AH
- Subjects
- Animals, Calcium metabolism, Cell Adhesion, Cell Movement, Cells, Cultured, Extracellular Traps, Female, Humans, Inflammation, Leukocytes cytology, Liver metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Peritonitis metabolism, Phosphorylation, Proto-Oncogene Proteins c-akt metabolism, RNA, Small Interfering metabolism, Signal Transduction, Factor XII metabolism, Neutrophils metabolism, Receptors, Urokinase Plasminogen Activator metabolism, Wound Healing
- Abstract
Coagulation factor XII (FXII) deficiency is associated with decreased neutrophil migration, but the mechanisms remain uncharacterized. Here, we examine how FXII contributes to the inflammatory response. In 2 models of sterile inflammation, FXII-deficient mice (F12-/-) had fewer neutrophils recruited than WT mice. We discovered that neutrophils produced a pool of FXII that is functionally distinct from hepatic-derived FXII and contributes to neutrophil trafficking at sites of inflammation. FXII signals in neutrophils through urokinase plasminogen activator receptor-mediated (uPAR-mediated) Akt2 phosphorylation at S474 (pAktS474). Downstream of pAkt2S474, FXII stimulation of neutrophils upregulated surface expression of αMβ2 integrin, increased intracellular calcium, and promoted extracellular DNA release. The sum of these activities contributed to neutrophil cell adhesion, migration, and release of neutrophil extracellular traps in a process called NETosis. Decreased neutrophil signaling in F12-/- mice resulted in less inflammation and faster wound healing. Targeting hepatic F12 with siRNA did not affect neutrophil migration, whereas WT BM transplanted into F12-/- hosts was sufficient to correct the neutrophil migration defect in F12-/- mice and restore wound inflammation. Importantly, these activities were a zymogen FXII function and independent of FXIIa and contact activation, highlighting that FXII has a sophisticated role in vivo that has not been previously appreciated.
- Published
- 2018
- Full Text
- View/download PDF
30. FSIP1 binds HER2 directly to regulate breast cancer growth and invasiveness.
- Author
-
Liu T, Zhang H, Sun L, Zhao D, Liu P, Yan M, Zaidi N, Izadmehr S, Gupta A, Abu-Amer W, Luo M, Yang J, Ou X, Wang Y, Bai X, Wang Y, New MI, Zaidi M, Yuen T, and Liu C
- Subjects
- Animals, Cell Line, Tumor, Cell Movement, Cell Proliferation, Epithelial-Mesenchymal Transition drug effects, Female, Gene Expression Profiling, Humans, MCF-7 Cells, Mice, Mice, Inbred BALB C, Mice, Nude, Neoplasm Invasiveness, Neoplasm Recurrence, Local genetics, Neoplasm Transplantation, Oligonucleotide Array Sequence Analysis, Protein Binding, Transcription Factors metabolism, Apoptosis, Breast Neoplasms metabolism, Carrier Proteins metabolism, Gene Expression Regulation, Neoplastic, Receptor, ErbB-2 metabolism, Seminal Plasma Proteins metabolism
- Abstract
Fibrous sheath interacting protein 1 (FSIP1), a spermatogenesis-related testicular antigen, is expressed in abundance in breast cancers, particularly in those overexpressing human epidermal growth factor receptor 2 (HER2); however, little is known about its role in regulating the growth and metastasis of breast cancer cells. We and others have shown previously that FSIP1 expression in breast cancer correlates positively with HER2-positivity, recurrence, and metastases and negatively with survival. Here, using coimmunoprecipitation and microscale thermophoresis, we find that FSIP1 binds to the intracellular domain of HER2 directly. We further show that shRNA-induced FSIP1 knockdown in SKBR3 and MCF-7 breast cancer cells inhibits proliferation, stimulates apoptosis, attenuates epithelial-mesenchymal transition, and impairs migration and invasiveness. Consistent with reduced proliferation and enhanced apoptosis, xenotransplantation of SKBR3 cells stably transfected with sh- FSIP1 into nu/nu mice results in reduced tumor volumes compared with sh-NC transplants. Furthermore, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) mapping using sh- FSIP1 gene signature yielded associations with extracellular matrix protein pathways, and a reduction in SNAI2 protein expression was confirmed on Western blot analysis. Complementarily, interrogation of the Connectivity Map using the same gene signature yielded, as top hits, chemicals known to inhibit epithelial-mesenchymal transition, including rapamycin, 17- N -allylamino-17-demethoxygeldanamycin, and LY294002. These compounds phenocopy the effects of sh- FSIP1 on SKBR3 cell viability. Thus, FSIP1 suppression limits oncogenesis and invasiveness in breast cancer cells and, considering its absence in most other tissues, including normal breast, may become a potential target for breast cancer therapy., Competing Interests: The authors declare no conflict of interest.
- Published
- 2017
- Full Text
- View/download PDF
31. Activation of tumor suppressor protein PP2A inhibits KRAS-driven tumor growth.
- Author
-
Sangodkar J, Perl A, Tohme R, Kiselar J, Kastrinsky DB, Zaware N, Izadmehr S, Mazhar S, Wiredja DD, O'Connor CM, Hoon D, Dhawan NS, Schlatzer D, Yao S, Leonard D, Borczuk AC, Gokulrangan G, Wang L, Svenson E, Farrington CC, Yuan E, Avelar RA, Stachnik A, Smith B, Gidwani V, Giannini HM, McQuaid D, McClinch K, Wang Z, Levine AC, Sears RC, Chen EY, Duan Q, Datt M, Haider S, Ma'ayan A, DiFeo A, Sharma N, Galsky MD, Brautigan DL, Ioannou YA, Xu W, Chance MR, Ohlmeyer M, and Narla G
- Subjects
- Animals, Antineoplastic Agents chemistry, Cell Line, Tumor, Cell Survival drug effects, Drug Resistance, Neoplasm, Enzyme Activation, Enzyme Activators chemistry, Humans, Male, Mice, Inbred BALB C, Mice, Nude, Mice, Transgenic, Protein Binding, Protein Phosphatase 2 chemistry, Signal Transduction, Tumor Burden, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Enzyme Activators pharmacology, Protein Phosphatase 2 metabolism, Proto-Oncogene Proteins p21(ras) genetics
- Abstract
Targeted cancer therapies, which act on specific cancer-associated molecular targets, are predominantly inhibitors of oncogenic kinases. While these drugs have achieved some clinical success, the inactivation of kinase signaling via stimulation of endogenous phosphatases has received minimal attention as an alternative targeted approach. Here, we have demonstrated that activation of the tumor suppressor protein phosphatase 2A (PP2A), a negative regulator of multiple oncogenic signaling proteins, is a promising therapeutic approach for the treatment of cancers. Our group previously developed a series of orally bioavailable small molecule activators of PP2A, termed SMAPs. We now report that SMAP treatment inhibited the growth of KRAS-mutant lung cancers in mouse xenografts and transgenic models. Mechanistically, we found that SMAPs act by binding to the PP2A Aα scaffold subunit to drive conformational changes in PP2A. These results show that PP2A can be activated in cancer cells to inhibit proliferation. Our strategy of reactivating endogenous PP2A may be applicable to the treatment of other diseases and represents an advancement toward the development of small molecule activators of tumor suppressor proteins.
- Published
- 2017
- Full Text
- View/download PDF
32. Blocking FSH induces thermogenic adipose tissue and reduces body fat.
- Author
-
Liu P, Ji Y, Yuen T, Rendina-Ruedy E, DeMambro VE, Dhawan S, Abu-Amer W, Izadmehr S, Zhou B, Shin AC, Latif R, Thangeswaran P, Gupta A, Li J, Shnayder V, Robinson ST, Yu YE, Zhang X, Yang F, Lu P, Zhou Y, Zhu LL, Oberlin DJ, Davies TF, Reagan MR, Brown A, Kumar TR, Epstein S, Iqbal J, Avadhani NG, New MI, Molina H, van Klinken JB, Guo EX, Buettner C, Haider S, Bian Z, Sun L, Rosen CJ, and Zaidi M
- Subjects
- Adipocytes drug effects, Adipocytes metabolism, Adipose Tissue drug effects, Adipose Tissue, Beige drug effects, Adipose Tissue, Beige metabolism, Adipose Tissue, White drug effects, Adipose Tissue, White metabolism, Animals, Antibodies immunology, Antibodies pharmacology, Diet, High-Fat adverse effects, Female, Follicle Stimulating Hormone, beta Subunit immunology, Haploinsufficiency, Male, Mice, Mitochondria drug effects, Mitochondria metabolism, Obesity drug therapy, Obesity prevention & control, Osteoporosis drug therapy, Ovariectomy, Oxygen Consumption drug effects, Receptors, FSH antagonists & inhibitors, Receptors, FSH genetics, Receptors, FSH metabolism, Uncoupling Protein 1 biosynthesis, Adipose Tissue metabolism, Adiposity drug effects, Follicle Stimulating Hormone, beta Subunit antagonists & inhibitors, Thermogenesis drug effects
- Abstract
Menopause is associated with bone loss and enhanced visceral adiposity. A polyclonal antibody that targets the β-subunit of the pituitary hormone follicle-stimulating hormone (Fsh) increases bone mass in mice. Here, we report that this antibody sharply reduces adipose tissue in wild-type mice, phenocopying genetic haploinsufficiency for the Fsh receptor gene Fshr. The antibody also causes profound beiging, increases cellular mitochondrial density, activates brown adipose tissue and enhances thermogenesis. These actions result from the specific binding of the antibody to the β-subunit of Fsh to block its action. Our studies uncover opportunities for simultaneously treating obesity and osteoporosis.
- Published
- 2017
- Full Text
- View/download PDF
33. Prostatic Acid Phosphatase Alters the RANKL/OPG System and Induces Osteoblastic Prostate Cancer Bone Metastases.
- Author
-
Kirschenbaum A, Izadmehr S, Yao S, O'Connor-Chapman KL, Huang A, Gregoriades EM, Yakar S, and Levine AC
- Subjects
- Acid Phosphatase genetics, Animals, Bone Neoplasms secondary, Cell Differentiation, Cell Line, Tumor, Cell Proliferation, Humans, Male, Mice, Mice, SCID, Osteoblasts pathology, Prostatic Neoplasms pathology, RNA, Small Interfering, Acid Phosphatase metabolism, Bone Neoplasms metabolism, Osteoblasts metabolism, Osteoprotegerin metabolism, Prostatic Neoplasms metabolism, RANK Ligand metabolism, Signal Transduction genetics
- Abstract
Prostate cancer (PCa) is unique in its tendency to produce osteoblastic (OB) bone metastases. There are no existing therapies that specifically target the OB phase that affects 90% of men with bone metastatic disease. Prostatic acid phosphatase (PAP) is secreted by PCa cells in OB metastases and increases OB growth, differentiation, and bone mineralization. The purpose of this study was to investigate whether PAP effects on OB bone metastases are mediated by autocrine and/or paracrine alterations in the receptor activator of nuclear factor κ-B (RANK)/RANK ligand (RANKL)/osteoprotegerin (OPG) system. To investigate whether PAP modulated these factors and altered the bone reaction, we knocked down PAP expression in VCaP cells and stably overexpressed PAP in PC3M cells, both derived from human PCa bone metastases. We show that knockdown of PAP in VCaP cells decreased OPG while increasing RANK/RANKL expression. Forced overexpression of PAP in PC3M cells had the inverse effect, increasing OPG while decreasing RANK/RANKL expression. Coculture of PCa cells with MC3T3 preosteoblasts also revealed a role for secretory PAP in OB-PCa cross talk. Reduced PAP expression in VCaP cells decreased MC3T3 proliferation and differentiation and reduced their OPG expression. PAP overexpression in PC3M cells altered the bone phenotype creating OB rather than osteolytic lesions in vivo using an intratibial model. These findings demonstrate that PAP secreted by PCa cells in OB bone metastases increases OPG and plays a critical role in the vicious cross talk between cancer and bone cells. These data suggest that inhibition of secretory PAP may be an effective strategy for PCa OB bone lesions.
- Published
- 2016
- Full Text
- View/download PDF
34. Clinical characteristics and outcomes of HIV-seropositive men treated with surgery for prostate cancer.
- Author
-
Izadmehr S, Leapman M, Hobbs AR, Katsigeorgis M, Nabizada-Pace F, Jazayeri SB, and Samadi DB
- Subjects
- Aged, Disease-Free Survival, HIV Seropositivity complications, Humans, Laparoscopy adverse effects, Laparoscopy methods, Male, Middle Aged, Neoplasm Grading, Neoplasm Staging, Outcome and Process Assessment, Health Care, Prostate pathology, Risk Assessment, Robotic Surgical Procedures adverse effects, Robotic Surgical Procedures methods, United States, Adenocarcinoma complications, Adenocarcinoma pathology, Adenocarcinoma surgery, Prostatectomy adverse effects, Prostatectomy methods, Prostatic Neoplasms complications, Prostatic Neoplasms pathology, Prostatic Neoplasms surgery
- Abstract
Purpose: The natural history and optimal management strategy for men with human immunodeficiency virus (HIV) and prostate cancer remain to be definitively characterized. This study was conducted to evaluate the clinical characteristics and outcomes of HIV-seropositive men treated with robotic-assisted radical laparoscopic prostatectomy for localized prostate cancer., Methods: After Institutional Review Board approval, a prospective database of 2175 operative cases of clinically localized prostate adenocarcinoma was reviewed. Thirteen patients were identified as HIV-positive. Tumor characteristics, operative outcomes, postoperative outcomes, histology (Gleason score), local invasion, biochemical recurrence, and surgical complications were compared with HIV-negative patients., Results: There were no preoperative demographic differences between the HIV-positive and HIV-negative patients. HIV-positive patients had higher prostate specific antigen (PSA) levels at time of diagnosis which was not statistically significant. However, HIV-positive patients had higher D'Amico risk assessment (p < 0.05). There was no postoperative complication. HIV-positive patients treated with robotic prostatectomy had similarly favorable perioperative and short-term biochemical recurrence-free survival outcomes., Conclusion: Our findings show that minimally invasive prostatectomy can be safely considered as a therapeutic option in otherwise eligible HIV-positive patients with clinically significant prostate cancer. Further research is necessary to outline a diagnostic and treatment guideline for HIV-positive men in detection and treatment of prostate cancer.
- Published
- 2016
- Full Text
- View/download PDF
35. Erratum to: Clinical characteristics and outcomes of HIV-seropositive men treated with surgery for prostate cancer.
- Author
-
Izadmehr S, Leapman M, Hobbs AR, Katsigeorgis M, Nabizada-Pace F, Jazayeri SB, and Samadi DB
- Published
- 2016
- Full Text
- View/download PDF
36. Corrigendum to "Reengineered tricyclic anti-cancer agents" [Bioorg. Med. Chem. 23 (2015) 6528-6534].
- Author
-
Kastrinsky DB, Sangodkar J, Zaware N, McClinch K, Farrington CC, Giannini HM, Izadmehr S, Dhawan NS, Narla G, and Ohlmeyer M
- Published
- 2015
- Full Text
- View/download PDF
37. Reengineered tricyclic anti-cancer agents.
- Author
-
Kastrinsky DB, Sangodkar J, Zaware N, Izadmehr S, Dhawan NS, Narla G, and Ohlmeyer M
- Subjects
- Animals, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Cell Line, Tumor, Cell Survival drug effects, Dibenzazepines chemistry, Heterocyclic Compounds, 3-Ring pharmacology, Heterocyclic Compounds, 3-Ring therapeutic use, Humans, Mice, Neoplasms drug therapy, Neoplasms metabolism, Phenothiazines chemistry, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction drug effects, Transplantation, Heterologous, Vesicular Monoamine Transport Proteins antagonists & inhibitors, Vesicular Monoamine Transport Proteins metabolism, Antineoplastic Agents chemistry, Heterocyclic Compounds, 3-Ring chemistry
- Abstract
The phenothiazine and dibenzazepine tricyclics are potent neurotropic drugs with a documented but underutilized anti-cancer side effect. Reengineering these agents (TFP, CPZ, CIP) by replacing the basic amine with a neutral polar functional group (e.g., RTC-1, RTC-2) abrogated their CNS effects as demonstrated by in vitro pharmacological assays and in vivo behavioral models. Further optimization generated several phenothiazines and dibenzazepines with improved anti-cancer potency, exemplified by RTC-5. This new lead demonstrated efficacy against a xenograft model of an EGFR driven cancer without the neurotropic effects exhibited by the parent molecules. Its effects were attributed to concomitant negative regulation of PI3K-AKT and RAS-ERK signaling., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
38. Repurposing of bisphosphonates for the prevention and therapy of nonsmall cell lung and breast cancer.
- Author
-
Stachnik A, Yuen T, Iqbal J, Sgobba M, Gupta Y, Lu P, Colaianni G, Ji Y, Zhu LL, Kim SM, Li J, Liu P, Izadmehr S, Sangodkar J, Scherer T, Mujtaba S, Galsky M, Gomez J, Epstein S, Buettner C, Bian Z, Zallone A, Aggarwal AK, Haider S, New MI, Sun L, Narla G, and Zaidi M
- Subjects
- Animals, Blotting, Western, Diphosphonates therapeutic use, Drug Repositioning methods, Female, Flow Cytometry, Humans, Immunohistochemistry, In Situ Nick-End Labeling, Mice, Mice, Inbred BALB C, Molecular Dynamics Simulation, Protein Binding, Signal Transduction drug effects, Tetrazolium Salts, Thiazoles, Tumor Stem Cell Assay, Breast Neoplasms drug therapy, Breast Neoplasms prevention & control, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung prevention & control, Diphosphonates pharmacology, ErbB Receptors antagonists & inhibitors
- Abstract
A variety of human cancers, including nonsmall cell lung (NSCLC), breast, and colon cancers, are driven by the human epidermal growth factor receptor (HER) family of receptor tyrosine kinases. Having shown that bisphosphonates, a class of drugs used widely for the therapy of osteoporosis and metastatic bone disease, reduce cancer cell viability by targeting HER1, we explored their potential utility in the prevention and therapy of HER-driven cancers. We show that bisphosphonates inhibit colony formation by HER1(ΔE746-A750)-driven HCC827 NSCLCs and HER1(wt)-expressing MB231 triple negative breast cancers, but not by HER(low)-SW620 colon cancers. In parallel, oral gavage with bisphosphonates of mice xenografted with HCC827 or MB231 cells led to a significant reduction in tumor volume in both treatment and prevention protocols. This result was not seen with mice harboring HER(low) SW620 xenografts. We next explored whether bisphosphonates can serve as adjunctive therapies to tyrosine kinase inhibitors (TKIs), namely gefitinib and erlotinib, and whether the drugs can target TKI-resistant NSCLCs. In silico docking, together with molecular dynamics and anisotropic network modeling, showed that bisphosphonates bind to TKIs within the HER1 kinase domain. As predicted from this combinatorial binding, bisphosphonates enhanced the effects of TKIs in reducing cell viability and driving tumor regression in mice. Impressively, the drugs also overcame erlotinib resistance acquired through the gatekeeper mutation T790M, thus offering an option for TKI-resistant NSCLCs. We suggest that bisphosphonates can potentially be repurposed for the prevention and adjunctive therapy of HER1-driven cancers.
- Published
- 2014
- Full Text
- View/download PDF
39. Bisphosphonates inactivate human EGFRs to exert antitumor actions.
- Author
-
Yuen T, Stachnik A, Iqbal J, Sgobba M, Gupta Y, Lu P, Colaianni G, Ji Y, Zhu LL, Kim SM, Li J, Liu P, Izadmehr S, Sangodkar J, Bailey J, Latif Y, Mujtaba S, Epstein S, Davies TF, Bian Z, Zallone A, Aggarwal AK, Haider S, New MI, Sun L, Narla G, and Zaidi M
- Subjects
- Anisotropy, Blotting, Western, Cell Line, Tumor, Crystallography, Diphosphonates metabolism, ErbB Receptors chemistry, ErbB Receptors metabolism, Fluorescence, Humans, Molecular Dynamics Simulation, Protein Binding, Protein Conformation, Tetrazolium Salts, Thiazoles, Antineoplastic Agents pharmacology, Apoptosis drug effects, Diphosphonates pharmacology, ErbB Receptors antagonists & inhibitors, Models, Molecular
- Abstract
Bisphosphonates are the most commonly prescribed medicines for osteoporosis and skeletal metastases. The drugs have also been shown to reduce cancer progression, but only in certain patient subgroups, suggesting that there is a molecular entity that mediates bisphosphonate action on tumor cells. Using connectivity mapping, we identified human epidermal growth factor receptors (human EGFR or HER) as a potential new molecular entity for bisphosphonate action. Protein thermal shift and cell-free kinase assays, together with computational modeling, demonstrated that N-containing bisphosphonates directly bind to the kinase domain of HER1/2 to cause a global reduction in downstream signaling. By doing so, the drugs kill lung, breast, and colon cancer cells that are driven by activating mutations or overexpression of HER1. Knocking down HER isoforms thus abrogates cell killing by bisphosphonates, establishing complete HER dependence and ruling out a significant role for other receptor tyrosine kinases or the enzyme farnesyl pyrophosphate synthase. Consistent with this finding, colon cancer cells expressing low levels of HER do not respond to bisphosphonates. The results suggest that bisphosphonates can potentially be repurposed for the prevention and therapy of HER family-driven cancers.
- Published
- 2014
- Full Text
- View/download PDF
40. Inducible nitric oxide synthase drives mTOR pathway activation and proliferation of human melanoma by reversible nitrosylation of TSC2.
- Author
-
Lopez-Rivera E, Jayaraman P, Parikh F, Davies MA, Ekmekcioglu S, Izadmehr S, Milton DR, Chipuk JE, Grimm EA, Estrada Y, Aguirre-Ghiso J, and Sikora AG
- Subjects
- Animals, Chick Embryo, Gene Knockdown Techniques, Humans, Melanoma metabolism, Nitric Oxide metabolism, Nitric Oxide Synthase Type II antagonists & inhibitors, Nitrosation drug effects, Nitrosation genetics, Protein Multimerization, Protein Processing, Post-Translational drug effects, Protein Processing, Post-Translational genetics, RNA, Small Interfering pharmacology, Signal Transduction drug effects, Signal Transduction genetics, Tuberous Sclerosis Complex 2 Protein, Tumor Cells, Cultured, Cell Proliferation drug effects, Melanoma pathology, Nitric Oxide Synthase Type II physiology, TOR Serine-Threonine Kinases metabolism, Tumor Suppressor Proteins metabolism
- Abstract
Melanoma is one of the cancers of fastest-rising incidence in the world. Inducible nitric oxide synthase (iNOS) is overexpressed in melanoma and other cancers, and previous data suggest that iNOS and nitric oxide (NO) drive survival and proliferation of human melanoma cells. However, specific mechanisms through which this occurs are poorly defined. One candidate is the PI3K-AKT-mTOR pathway, which plays a major role in proliferation, angiogenesis, and metastasis of melanoma and other cancers. We used the chick embryo chorioallantoic membrane (CAM) assay to test the hypothesis that melanoma growth is regulated by iNOS-dependent mTOR pathway activation. Both pharmacologic inhibition and siRNA-mediated gene silencing of iNOS suppressed melanoma proliferation and in vivo growth on the CAM in human melanoma models. This was associated with strong downregulation of mTOR pathway activation by Western blot analysis of p-mTOR, p70 ribosomal S6 kinase (p-P70S6K), p-S6RP, and p-4EBP1. iNOS expression and NO were associated with reversible nitrosylation of tuberous sclerosis complex (TSC) 2, and inhibited dimerization of TSC2 with its inhibitory partner TSC1, enhancing GTPase activity of its target Ras homolog enriched in brain (Rheb), a critical activator of mTOR signaling. Immunohistochemical analysis of tumor specimens from stage III melanoma patients showed a significant correlation between iNOS expression levels and expression of the mTOR pathway members. Exogenously supplied NO was also sufficient to reverse the mTOR pathway inhibition by the B-Raf inhibitor vemurafenib. In summary, covalent modification of TSC2 by iNOS-derived NO is associated with impaired TSC2/TSC1 dimerization, mTOR pathway activation, and proliferation of human melanoma. This model is consistent with the known association of iNOS overexpression and poor prognosis in melanoma and other cancers., (©2014 AACR.)
- Published
- 2014
- Full Text
- View/download PDF
41. KLF6-SV1 drives breast cancer metastasis and is associated with poor survival.
- Author
-
Hatami R, Sieuwerts AM, Izadmehr S, Yao Z, Qiao RF, Papa L, Look MP, Smid M, Ohlssen J, Levine AC, Germain D, Burstein D, Kirschenbaum A, DiFeo A, Foekens JA, and Narla G
- Subjects
- Adult, Aged, Animals, Cell Movement, Cell Proliferation, Cell Survival, Disease Progression, Epithelial-Mesenchymal Transition genetics, Female, Gene Expression Regulation, Neoplastic, Humans, Kruppel-Like Factor 6, Kruppel-Like Transcription Factors metabolism, Mice, Middle Aged, Multivariate Analysis, Neoplasm Invasiveness, Neoplasm Metastasis genetics, Nuclear Proteins, Phenotype, Prognosis, Proportional Hazards Models, Proto-Oncogene Proteins metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Survival Analysis, Twist-Related Protein 1, Xenograft Model Antitumor Assays, Alternative Splicing genetics, Breast Neoplasms genetics, Breast Neoplasms pathology, Kruppel-Like Transcription Factors genetics, Proto-Oncogene Proteins genetics
- Abstract
Metastasis is the major cause of cancer mortality. A more thorough understanding of the mechanisms driving this complex multistep process will aid in the identification and characterization of therapeutically targetable genetic drivers of disease progression. We demonstrate that KLF6-SV1, an oncogenic splice variant of the KLF6 tumor suppressor gene, is associated with increased metastatic potential and poor survival in a cohort of 671 lymph node-negative breast cancer patients. KLF6-SV1 overexpression in mammary epithelial cell lines resulted in an epithelial-to-mesenchymal-like transition and drove aggressive multiorgan metastatic disease in multiple in vivo models. Additionally, KLF6-SV1 loss-of-function studies demonstrated reversion to an epithelial and less invasive phenotype. Combined, these findings implicate KLF6-SV1 as a key driver of breast cancer metastasis that distinguishes between indolent and lethal early-stage disease and provides a potential therapeutic target for invasive breast cancer.
- Published
- 2013
- Full Text
- View/download PDF
42. Targeting the FOXO1/KLF6 axis regulates EGFR signaling and treatment response.
- Author
-
Sangodkar J, Dhawan NS, Melville H, Singh VJ, Yuan E, Rana H, Izadmehr S, Farrington C, Mazhar S, Katz S, Albano T, Arnovitz P, Okrent R, Ohlmeyer M, Galsky M, Burstein D, Zhang D, Politi K, Difeo A, and Narla G
- Subjects
- Active Transport, Cell Nucleus drug effects, Adenocarcinoma drug therapy, Adenocarcinoma pathology, Adenocarcinoma of Lung, Animals, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Cell Line, Tumor, Drug Resistance, Neoplasm, Drug Synergism, Enzyme Activation, ErbB Receptors antagonists & inhibitors, Erlotinib Hydrochloride, Female, Forkhead Box Protein O1, Forkhead Transcription Factors genetics, Gene Expression Regulation, Neoplastic, Humans, Kruppel-Like Factor 6, Kruppel-Like Transcription Factors genetics, Lung Neoplasms drug therapy, Lung Neoplasms pathology, Mice, Mice, Inbred BALB C, Mice, Nude, Mutation, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins c-akt metabolism, Quinazolines pharmacology, Quinazolines therapeutic use, Real-Time Polymerase Chain Reaction, Signal Transduction, Transcription, Genetic, Trifluoperazine pharmacology, Tumor Burden drug effects, Xenograft Model Antitumor Assays, Adenocarcinoma metabolism, ErbB Receptors metabolism, Forkhead Transcription Factors metabolism, Kruppel-Like Transcription Factors metabolism, Lung Neoplasms metabolism, Proto-Oncogene Proteins metabolism
- Abstract
EGFR activation is both a key molecular driver of disease progression and the target of a broad class of molecular agents designed to treat advanced cancer. Nevertheless, resistance develops through several mechanisms, including activation of AKT signaling. Though much is known about the specific molecular lesions conferring resistance to anti-EGFR-based therapies, additional molecular characterization of the downstream mediators of EGFR signaling may lead to the development of new classes of targeted molecular therapies to treat resistant disease. We identified a transcriptional network involving the tumor suppressors Krüppel-like factor 6 (KLF6) and forkhead box O1 (FOXO1) that negatively regulates activated EGFR signaling in both cell culture and in vivo models. Furthermore, the use of the FDA-approved drug trifluoperazine hydrochloride (TFP), which has been shown to inhibit FOXO1 nuclear export, restored sensitivity to AKT-driven erlotinib resistance through modulation of the KLF6/FOXO1 signaling cascade in both cell culture and xenograft models of lung adenocarcinoma. Combined, these findings define a novel transcriptional network regulating oncogenic EGFR signaling and identify a class of FDA-approved drugs as capable of restoring chemosensitivity to anti-EGFR-based therapy for the treatment of metastatic lung adenocarcinoma.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.