86 results on '"Pitts TM"'
Search Results
2. Abstract P2-06-15: Rational combination of Wee1 and BCL-2 inhibition in preclinical models of triple-negative breast cancer
- Author
-
Simmons, DM, primary, Tse, TE, additional, Dailey, K, additional, Hartman, SJ, additional, Bagsby, S, additional, Pitts, TM, additional, Tentler, JJ, additional, and Diamond, JR, additional
- Published
- 2019
- Full Text
- View/download PDF
3. Abstract P5-21-16: Preclinical studies of RX-5902, a beta-catenin modulator in triple negative breast cancer
- Author
-
Tentler, JJ, primary, Frank, JG, additional, Kim, DJ, additional, George, C, additional, Lee, YB, additional, Ely, B, additional, Tan, AC, additional, Kim, J, additional, Pitts, TM, additional, Capasso, A, additional, Dailey, KL, additional, Eckhardt, G, additional, and Diamond, JR, additional
- Published
- 2018
- Full Text
- View/download PDF
4. Abstract PD3-16: Clinical safety and efficacy of the aurora and angiogenic kinase inhibitor ENMD-2076 in previously treated, locally advanced or metastatic triple-negative breast cancer
- Author
-
Diamond, JR, primary, Eckhardt, SG, additional, Pitts, TM, additional, van Bokhoven, A, additional, Aisner, D, additional, Gustafson, DL, additional, Capasso, A, additional, Elias, AD, additional, Storniolo, AM, additional, Schneider, BP, additional, Gao, D, additional, Tentler, JJ, additional, Borges, VF, additional, and Miller, KD, additional
- Published
- 2018
- Full Text
- View/download PDF
5. Preclinical Activity of the Rational Combination of Selumetinib (AZD6244) in Combination with Vorinostat in KRAS-Mutant Colorectal Cancer Models
- Author
-
Morelli MP, Tentler JJ, Kulikowski GN, Tan AC, Bradshaw Pierce EL, Pitts TM, Brown AM, Nallapareddy S, Arcaroli JJ, Serkova NJ, Hidalgo M, Eckhardt S.G., CIARDIELLO, Fortunato, Morelli, Mp, Tentler, Jj, Kulikowski, Gn, Tan, Ac, Bradshaw Pierce, El, Pitts, Tm, Brown, Am, Nallapareddy, S, Arcaroli, Jj, Serkova, Nj, Hidalgo, M, Ciardiello, Fortunato, and Eckhardt, S. G.
- Published
- 2012
6. Abstract P2-09-06: The role of p53 family tumor suppressors in mediating response to aurora kinase inhibition in triple-negative breast cancer
- Author
-
Diamond, JR, primary, Tan, AC, additional, Ionkina, AA, additional, Newton, TP, additional, Pitts, TM, additional, Eckhardt, SG, additional, and Tentler, JJ, additional
- Published
- 2013
- Full Text
- View/download PDF
7. P3-01-08: In Vitro and In Vivo Antitumor Activity of the Aurora and Angiogenic Kinase Inhibitor ENMD-2076 in Triple-Negative Breast Cancer Models.
- Author
-
Diamond, JR, primary, Eckhardt, SG, additional, Tan, AC, additional, Selby, HM, additional, Newton, TP, additional, Pitts, TM, additional, Bray, MR, additional, Fletcher, GC, additional, and Tentler, JJ, additional
- Published
- 2011
- Full Text
- View/download PDF
8. Targeting vascular endothelial growth factor receptor-1 and -3 with cediranib (AZD2171): effects on migration and invasion of gastrointestinal cancer cell lines
- Author
-
Amy M. Brown, M. Pia Morelli, Anderson J. Ryan, Todd M. Pitts, Fortunato Ciardiello, John J. Tentler, Juliane M. Jürgensmeier, S. Gail Eckhardt, Morelli, Mp, Brown, Am, Pitts, Tm, Tentler, Jj, Ciardiello, Fortunato, Ryan, A, Jürgensmeier, Jm, and Eckhardt, Sg
- Subjects
Cancer Research ,medicine.medical_specialty ,Cell ,Blotting, Western ,Antineoplastic Agents ,Enzyme-Linked Immunosorbent Assay ,Biology ,Article ,Flow cytometry ,Cediranib ,chemistry.chemical_compound ,Cell Movement ,Internal medicine ,Cell Line, Tumor ,medicine ,Humans ,Secretion ,Neoplasm Invasiveness ,Autocrine signalling ,Cell Proliferation ,Gastrointestinal Neoplasms ,Vascular Endothelial Growth Factor Receptor-1 ,medicine.diagnostic_test ,integumentary system ,Cell growth ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Flow Cytometry ,Vascular Endothelial Growth Factor Receptor-3 ,Vascular endothelial growth factor ,Endocrinology ,medicine.anatomical_structure ,Oncology ,chemistry ,Cell culture ,embryonic structures ,Cancer research ,Quinazolines ,cardiovascular system ,medicine.drug - Abstract
The effect of vascular endothelial growth factor (VEGF) ligands and cediranib on tumor cell proliferation, migration, and invasion was determined. It has recently been suggested that autocrine signaling through the VEGF receptor (VEGFR) pathway may play a role in tumor cell survival, invasion, and migration. The purpose of the present study was to determine the expression of VEGFRs and VEGFR ligands in a panel of gastrointestinal carcinoma cells. Additionally, we evaluated the effects of VEGF autocrine signaling on tumor cell proliferation, migration, and invasion utilizing cediranib (AZD2171), a pan-VEGFR inhibitor. Five colorectal, three pancreatic, and two hepatocellular carcinoma cell lines were screened for VEGFR and VEGF expression by several methods. Expression of VEGFR-1 and VEGFR-3 was cell line–dependent, whereas VEGFR-2 was not detected. Secretion of VEGF-A was detected in the supernatants of all cell lines whereas VEGF-C secretion was detected in the Panc-1, MiaPaca2, and Hep1 cells only. Tumor cells showed increased migratory activity, but not proliferation, when stimulated with VEGFs. The pan-VEGFR inhibitor cediranib (100 nmol/L) inhibited tumor cell migration and invasion, with no effects on proliferation. Cediranib decreased VEGFR-1 and VEGFR-3 phosphorylation as well as activation of downstream effectors. VEGFR-1 and VEGFR-3 expression was detected in all the gastrointestinal carcinoma cells evaluated. Although activation of the VEGF pathway did not affect cell proliferation, our data indicate that this pathway seems to play a role in tumor cell migration and invasion in these cell lines. Therefore, inhibition of VEGFR by cediranib may represent a clinically relevant treatment option for gastrointestinal tumors. [Mol Cancer Ther 2009;8(9):2546–58]
- Published
- 2016
9. Expanding the landscape of oncogenic drivers and treatment options in acral and mucosal melanomas by targeted genomic profiling.
- Author
-
Turner JA, Van Gulick RJ, Robinson WA, Mughal T, Tobin RP, MacBeth ML, Holman B, Classon A, Bagby SM, Yacob BW, Hartman SJ, Silverman I, Vorwald VM, Gorden N, Gonzalez R, Gay LM, Ali SM, Benson A, Miller VA, Ross JS, Pitts TM, Rioth MJ, Lewis KD, Medina T, McCarter MD, Gonzalez R, and Couts KL
- Subjects
- Humans, Male, Female, Middle Aged, Animals, Aged, Mice, Cell Line, Tumor, Adult, Genomics methods, Triazoles therapeutic use, Triazoles pharmacology, Azepines pharmacology, Azepines therapeutic use, Molecular Targeted Therapy methods, Gene Expression Profiling methods, Melanoma genetics, Melanoma drug therapy, Melanoma pathology, Skin Neoplasms genetics, Skin Neoplasms drug therapy, Skin Neoplasms pathology, Dasatinib therapeutic use, Dasatinib pharmacology, Mucous Membrane pathology, Mucous Membrane drug effects, Mucous Membrane metabolism
- Abstract
Despite advancements in treating cutaneous melanoma, patients with acral and mucosal (A/M) melanomas still have limited therapeutic options and poor prognoses. We analyzed 156 melanomas (101 cutaneous, 28 acral, and 27 mucosal) using the Foundation One cancer-gene specific clinical testing platform and identified new, potentially targetable genomic alterations (GAs) in specific anatomic sites of A/M melanomas. Using novel pre-clinical models of A/M melanoma, we demonstrate that several GAs and corresponding oncogenic pathways associated with cutaneous melanomas are similarly targetable in A/M melanomas. Other alterations, including MYC and CRKL amplifications, were unique to A/M melanomas and susceptible to indirect targeting using the BRD4 inhibitor JQ1 or Src/ABL inhibitor dasatinib, respectively. We further identified new, actionable A/M-specific alterations, including an inactivating NF2 fusion in a mucosal melanoma responsive to dasatinib in vivo. Our study highlights new molecular differences between cutaneous and A/M melanomas, and across different anatomic sites within A/M, which may change clinical testing and treatment paradigms for these rare melanomas., (© 2024 UICC.)
- Published
- 2024
- Full Text
- View/download PDF
10. A Phase II Study of Potentiation of Pembrolizumab with Binimetinib and Bevacizumab in Refractory Microsatellite-Stable Colorectal Cancer.
- Author
-
Lentz RW, Friedrich TJ, Blatchford PJ, Jordan KR, Pitts TM, Robinson HR, Davis SL, Kim SS, Leal AD, Lee MR, Waring MRN, Martin AC, Dominguez ATA, Bagby SM, Hartman SJ, Eckhardt SG, Messersmith WA, and Lieu CH
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Adult, Drug Resistance, Neoplasm genetics, Microsatellite Repeats, Treatment Outcome, Bevacizumab administration & dosage, Bevacizumab therapeutic use, Bevacizumab adverse effects, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Colorectal Neoplasms mortality, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized adverse effects, Benzimidazoles administration & dosage, Benzimidazoles therapeutic use, Benzimidazoles adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects
- Abstract
Purpose: In this single-institution phase II investigator-initiated study, we assessed the ability of MAPK and VEGF pathway blockade to overcome resistance to immunotherapy in microsatellite-stable metastatic colorectal cancer (MSS mCRC)., Patients and Methods: Patients with MSS, BRAF wild-type mCRC who progressed on ≥2 prior lines of therapy received pembrolizumab, binimetinib, and bevacizumab until disease progression or unacceptable toxicity. After a safety run-in, patients were randomized to a 7-day run-in of binimetinib or simultaneous initiation of all study drugs, to explore whether MEK inhibition may increase tumor immunogenicity. The primary endpoint was objective response rate (ORR) in all patients combined (by Response Evaluation Criteria in Solid Tumors v1.1)., Results: Fifty patients received study drug treatment; 54% were male with a median age of 55 years (range, 31-79). The primary endpoint, ORR, was 12.0% [95% confidence interval (CI) 4.5%-24.3%], which was not statistically different than the historical control data of 5% (P = 0.038, exceeding prespecified threshold of 0.025). The disease control rate was 70.0% (95% CI, 55.4%-82.1%), the median progression-free survival 5.9 months (95% CI, 4.2-8.7 months), and the median overall survival 9.3 months (95% CI, 6.7-12.2 months). No difference in efficacy was observed between the randomized cohorts. Grade 3 and 4 adverse events were observed in 56% and 8% of patients, respectively; the most common were rash (12%) and increased aspartate aminotransferase (12%)., Conclusions: Pembrolizumab, binimetinib, and bevacizumab failed to meet its primary endpoint of higher ORR compared with historical control data, demonstrated a high disease control rate, and demonstrated acceptable tolerability in refractory MSS mCRC., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
11. Endogenous retroviruses mediate transcriptional rewiring in response to oncogenic signaling in colorectal cancer.
- Author
-
Ivancevic A, Simpson DM, Joyner OM, Bagby SM, Nguyen LL, Bitler BG, Pitts TM, and Chuong EB
- Subjects
- Humans, Enhancer Elements, Genetic, Cell Line, Tumor, Transcription, Genetic, Animals, Carcinogenesis genetics, Gene Regulatory Networks, Colorectal Neoplasms genetics, Colorectal Neoplasms virology, Colorectal Neoplasms pathology, Colorectal Neoplasms metabolism, Endogenous Retroviruses genetics, Gene Expression Regulation, Neoplastic, Signal Transduction
- Abstract
Cancer cells exhibit rewired transcriptional regulatory networks that promote tumor growth and survival. However, the mechanisms underlying the formation of these pathological networks remain poorly understood. Through a pan-cancer epigenomic analysis, we found that primate-specific endogenous retroviruses (ERVs) are a rich source of enhancers displaying cancer-specific activity. In colorectal cancer and other epithelial tumors, oncogenic MAPK/AP1 signaling drives the activation of enhancers derived from the primate-specific ERV family LTR10. Functional studies in colorectal cancer cells revealed that LTR10 elements regulate tumor-specific expression of multiple genes associated with tumorigenesis, such as ATG12 and XRCC4 . Within the human population, individual LTR10 elements exhibit germline and somatic structural variation resulting from a highly mutable internal tandem repeat region, which affects AP1 binding activity. Our findings reveal that ERV-derived enhancers contribute to transcriptional dysregulation in response to oncogenic signaling and shape the evolution of cancer-specific regulatory networks.
- Published
- 2024
- Full Text
- View/download PDF
12. Examination of Wnt signaling as a therapeutic target for pancreatic ductal adenocarcinoma (PDAC) using a pancreatic tumor organoid library (PTOL).
- Author
-
Hawkins HJ, Yacob BW, Brown ME, Goldstein BR, Arcaroli JJ, Bagby SM, Hartman SJ, Macbeth M, Goodspeed A, Danhorn T, Lentz RW, Lieu CH, Leal AD, Messersmith WA, Dempsey PJ, and Pitts TM
- Subjects
- Animals, Mice, Humans, Gemcitabine, Wnt Signaling Pathway, Deoxycytidine pharmacology, Deoxycytidine therapeutic use, Mice, Nude, Cell Proliferation, Cell Line, Tumor, Paclitaxel pharmacology, Paclitaxel therapeutic use, Organoids metabolism, Xenograft Model Antitumor Assays, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal metabolism, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents at advanced stages and is refractory to most treatment modalities. Wnt signaling activation plays a critical role in proliferation and chemotherapeutic resistance. Minimal media conditions, growth factor dependency, and Wnt dependency were determined via Wnt inhibition for seven patient derived organoids (PDOs) derived from pancreatic tumor organoid libraries (PTOL). Organoids demonstrating response in vitro were assessed in vivo using patient-derived xenografts. Wnt (in)dependent gene signatures were identified for each organoid. Panc269 demonstrated a trend of reduced organoid growth when treated with ETC-159 in combination with paclitaxel or gemcitabine as compared with chemotherapy or ETC-159 alone. Panc320 demonstrated a more pronounced anti-proliferative effect in the combination of ETC-159 and paclitaxel but not with gemcitabine. Panc269 and Panc320 were implanted into nude mice and treated with ETC-159, paclitaxel, and gemcitabine as single agents and in combination. The combination of ETC-159 and paclitaxel demonstrated an anti-tumor effect greater than ETC-159 alone. Extent of combinatory treatment effect were observed to a lesser extent in the Panc320 xenograft. Wnt (in)dependent gene signatures of Panc269 and 320 were consistent with the phenotypes displayed. Gene expression of several key Wnt genes assessed via RT-PCR demonstrated notable fold change following treatment in vivo. Each pancreatic organoid demonstrated varied niche factor dependencies, providing an avenue for targeted therapy, supported through growth analysis following combinatory treatment of Wnt inhibitor and standard chemotherapy in vitro. The clinical utilization of this combinatory treatment modality in pancreatic cancer PDOs has thus far been supported in our patient-derived xenograft models treated with Wnt inhibitor plus paclitaxel or gemcitabine. Gene expression analysis suggests there are key Wnt genes that contribute to the Wnt (in)dependent phenotypes of pancreatic tumors, providing plausible mechanistic explanation for Wnt (in)dependency and susceptibility or resistance to treatment on the genotypic level., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2024 Hawkins et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2024
- Full Text
- View/download PDF
13. A Phase Ib Expansion Cohort Evaluating Aurora A Kinase Inhibitor Alisertib and Dual TORC1/2 Inhibitor Sapanisertib in Patients with Advanced Solid Tumors.
- Author
-
Davis SL, Messersmith WA, Purcell WT, Lam ET, Corr BR, Leal AD, Lieu CH, O'Bryant CL, Smoots SG, Dus ED, Jordan KR, Serkova NJ, Pitts TM, and Diamond JR
- Abstract
Background: This study further evaluated the safety and efficacy of the combination of alisertib and sapanisertib in an expansion cohort of patients, including a subset of patients with refractory pancreatic adenocarcinoma, with further evaluation of the pharmacodynamic characteristics of combination therapy., Methods: Twenty patients with refractory solid tumors and 11 patients with pancreatic adenocarcinoma were treated at the recommended phase 2 dose of alisertib and sapanisertib. Adverse events and disease response were assessed. Patients in the expansion cohort were treated with a 7-day lead-in of either alisertib or sapanisertib prior to combination therapy, with tumor tissue biopsy and serial functional imaging performed for correlative analysis., Results: Toxicity across treatment groups was overall similar to prior studies. One partial response to treatment was observed in a patient with ER positive breast cancer, and a patient with pancreatic cancer experienced prolonged stable disease. In an additional cohort of pancreatic cancer patients, treatment response was modest. Correlative analysis revealed variability in markers of apoptosis and immune cell infiltrate according to lead-in therapy and response., Conclusions: Dual targeting of Aurora A kinase and mTOR resulted in marginal clinical benefit in a population of patients with refractory solid tumors, including pancreatic adenocarcinoma, though individual patients experienced significant response to therapy. Correlatives indicate apoptotic response and tumor immune cell infiltrate may affect clinical outcomes.
- Published
- 2024
- Full Text
- View/download PDF
14. Overcoming doxorubicin resistance in triple-negative breast cancer using the class I-targeting HDAC inhibitor bocodepsin/OKI-179 to promote apoptosis.
- Author
-
Smoots SG, Schreiber AR, Jackson MM, Bagby SM, Dominguez ATA, Dus ED, Binns CA, MacBeth M, Whitty PA, Diamond JR, and Pitts TM
- Subjects
- Humans, Tumor Suppressor Protein p53 genetics, Doxorubicin pharmacology, Doxorubicin therapeutic use, Apoptosis, RNA, Small Interfering, Histone Deacetylase Inhibitors pharmacology, Histone Deacetylase Inhibitors therapeutic use, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms metabolism
- Abstract
Background: Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype with a poor prognosis. Doxorubicin is part of standard curative therapy for TNBC, but chemotherapy resistance remains an important clinical challenge. Bocodepsin (OKI-179) is a small molecule class I histone deacetylase (HDAC) inhibitor that promotes apoptosis in TNBC preclinical models. The purpose of this study was to investigate the combination of bocodepsin and doxorubicin in preclinical TNBC models and evaluate the impact on terminal cell fate, including apoptosis and senescence., Methods: TNBC cell lines were treated with doxorubicin and CellTiter-Glo was used to assess proliferation and determine doxorubicin sensitivity. Select cell lines were treated with OKI-005 (in vitro version of bocodepsin) and doxorubicin and assessed for proliferation, apoptosis as measured by Annexin V/PI, and cell cycle by flow cytometry. Immunoblotting was used to assess changes in mediators of apoptosis, cell cycle arrest, and senescence. Senescence was measured by the senescence-associated β-galactosidase assay. An MDA-MB-231 xenograft in vivo model was treated with bocodepsin, doxorubicin, or the combination and assessed for inhibition of tumor growth. shRNA knockdown of p53 was performed in the CAL-51 cell line and proliferation, apoptosis and senescence were assessed in response to combination treatment., Results: OKI-005 and doxorubicin resulted in synergistic antiproliferative activity in TNBC cells lines regardless of p53 mutation status. The combination led to increased apoptosis and decreased senescence. In vivo, the combination resulted in increased tumor growth inhibition compared to either single agent. shRNA knock-down of p53 led to increased doxorubicin-induced senescence that was decreased with the addition of OKI-005 in vitro., Conclusion: The addition of bocodepsin to doxorubicin resulted in synergistic antiproliferative activity in vitro, improved tumor growth inhibition in vivo, and promotion of apoptosis which makes this a promising combination to overcome doxorubicin resistance in TNBC. Bocodepsin is currently in clinical development and has a favorable toxicity profile compared to other HDAC inhibitors supporting the feasibility of evaluating this combination in patients with TNBC., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
15. Population-level comparisons of gene regulatory networks modeled on high-throughput single-cell transcriptomics data.
- Author
-
Osorio D, Capasso A, Eckhardt SG, Giri U, Somma A, Pitts TM, Lieu CH, Messersmith WA, Bagby SM, Singh H, Das J, Sahni N, Yi SS, and Kuijjer ML
- Subjects
- Humans, Gene Expression Profiling, Algorithms, RNA, Gene Regulatory Networks, Gene Expression Regulation
- Abstract
Single-cell technologies enable high-resolution studies of phenotype-defining molecular mechanisms. However, data sparsity and cellular heterogeneity make modeling biological variability across single-cell samples difficult. Here we present SCORPION, a tool that uses a message-passing algorithm to reconstruct comparable gene regulatory networks from single-cell/nuclei RNA-sequencing data that are suitable for population-level comparisons by leveraging the same baseline priors. Using synthetic data, we found that SCORPION outperformed 12 existing gene regulatory network reconstruction techniques. Using supervised experiments, we show that SCORPION can accurately identify differences in regulatory networks between wild-type and transcription factor-perturbed cells. We demonstrate SCORPION's scalability to population-level analyses using a single-cell RNA-sequencing atlas containing 200,436 cells from colorectal cancer and adjacent healthy tissues. The differences between tumor regions detected by SCORPION are consistent across multiple cohorts as well as with our understanding of disease progression, and elucidate phenotypic regulators that may impact patient survival., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
16. Pancreatic Ductal Adenocarcinoma Cells Regulate NLRP3 Activation to Generate a Tolerogenic Microenvironment.
- Author
-
Amo-Aparicio J, Dominguez A, Atif SM, Dinarello A, Azam T, Alula KM, Piper M, Lieu CH, Lentz RW, Leal AD, Bagby SM, Messersmith WA, Karam SD, Dinarello CA, Pitts TM, and Marchetti C
- Abstract
Defining feature of pancreatic ductal adenocarcinoma (PDAC) that participates in the high mortality rate and drug resistance is the immune-tolerant microenvironment which enables tumors to progress unabated by adaptive immunity. In this study, we report that PDAC cells release CSF-1 to induce nucleotide-binding domain, leucine-rich containing family, pyrin domain-containing-3 (NLRP3) activation in myeloid cells. Increased NLRP3 expression was found in the pancreas of patients with PDAC when compared with normal pancreas which correlated with the formation of the NLRP3 inflammasome. Using human primary cells and an orthotopic PDAC mouse model, we show that NLRP3 activation is responsible for the maturation and release of the inflammatory cytokine IL1β which selectively drives Th2-type inflammation via COX2/PGE2 induction. As a result of this inflammation, primary tumors were characterized by reduced cytotoxic CD8+ T-cell activation and increased tumor expansion. Genetic deletion and pharmacologic inhibition of NLRP3 enabled the development of Th1 immunity, increased intratumoral levels of IL2, CD8+ T cell–mediated tumor suppression, and ultimately limited tumor growth. In addition, we observed that NLRP3 inhibition in combination with gemcitabine significantly increased the efficacy of the chemotherapy. In conclusion, this study provides a mechanism by which tumor-mediated NLRP3 activation exploits a distinct adaptive immunity response that facilitates tumor escape and progression. Considering the ability to block NLRP3 activity with safe and small orally active molecules, this protein represents a new promising target to improve the limited therapeutic options in PDAC., Significant: This study provides novel molecular insights on how PDAC cells exploit NLRP3 activation to suppress CD8 T-cell activation. From a translational perspective, we demonstrate that the combination of gemcitabine with the orally active NLRP3 inhibitor OLT1177 increases the efficacy of monotherapy., (© 2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
17. Testing Cancer Immunotherapeutics in a Humanized Mouse Model Bearing Human Tumors.
- Author
-
Lanis JM, Lewis MS, Strassburger H, Larsen K, Bagby SM, Dominguez ATA, Marín-Jiménez JA, Pelanda R, Pitts TM, and Lang J
- Subjects
- Humans, Animals, Mice, Xenograft Model Antitumor Assays, Mice, Inbred NOD, Transplantation, Heterologous, Immunotherapy methods, Disease Models, Animal, Tumor Microenvironment, Neoplasms pathology
- Abstract
Reversing the immunosuppressive nature of the tumor microenvironment is critical for the successful treatment of cancers with immunotherapy drugs. Murine cancer models are extremely limited in their diversity and suffer from poor translation to the clinic. To serve as a more physiological preclinical model for immunotherapy studies, this protocol has been developed to evaluate the treatment of human tumors in a mouse reconstituted with a human immune system. This unique protocol demonstrates the development of human immune system (HIS, "humanized") mice, followed by implantation of a human tumor, either a cell-line derived xenograft (CDX) or a patient derived xenograft (PDX). HIS mice are generated by injecting CD34+ human hematopoietic stem cells isolated from umbilical cord blood into neonatal BRGS (BALB/c Rag2
-/- IL2RγC-/- NODSIRPα ) highly immunodeficient mice that are also capable of accepting a xenogeneic tumor. The importance of the kinetics and characteristics of the human immune system development and tumor implantation is emphasized. Finally, an in-depth evaluation of the tumor microenvironment using flow cytometry is described. In numerous studies using this protocol, it was found that the tumor microenvironment of individual tumors is recapitulated in HIS-PDX mice; "hot" tumors exhibit large immune infiltration while "cold" tumors do not. This model serves as a testing ground for combination immunotherapies for a wide range of human tumors and represents an important tool in the quest for personalized medicine.- Published
- 2022
- Full Text
- View/download PDF
18. Cabozantinib sensitizes microsatellite stable colorectal cancer to immune checkpoint blockade by immune modulation in human immune system mouse models.
- Author
-
Lang J, Leal AD, Marín-Jiménez JA, Hartman SJ, Shulman J, Navarro NM, Lewis MS, Capasso A, Bagby SM, Yacob BW, MacBeth M, Freed BM, Eckhardt SG, Jordan K, Blatchford PJ, Pelanda R, Lieu CH, Messersmith WA, and Pitts TM
- Abstract
Immune checkpoint inhibitors have been found to be effective in metastatic MSI-high colorectal cancers (CRC), however, have no efficacy in microsatellite stable (MSS) cancers, which comprise the majority of mCRC cases. Cabozantinib is a small molecule multi-tyrosine kinase inhibitor that is FDA approved in advanced renal cell, medullary thyroid, and hepatocellular carcinoma. Using Human Immune System (HIS) mice, we tested the ability of cabozantinib to prime MSS-CRC tumors to enhance the potency of immune checkpoint inhibitor nivolumab. In four independent experiments, we implanted distinct MSS-CRC patient-derived xenografts (PDXs) into the flanks of humanized BALB/c-Rag2
null Il2rγnull SirpαNOD (BRGS) mice that had been engrafted with human hematopoietic stem cells at birth. For each PDX, HIS-mice cohorts were treated with vehicle, nivolumab, cabozantinib, or the combination. In three out of the four models, the combination had a lower tumor growth rate compared to vehicle or nivolumab-treated groups. Furthermore, interrogation of the HIS in immune organs and tumors by flow cytometry revealed increased Granzyme B+, TNFα+ and IFNγ+ CD4+ T cells among the human tumor infiltrating leukocytes (TIL) that correlated with reduced tumor growth in the combination-treated HIS-mice. Notably, slower growth correlated with increased expression of the CD4+ T cell ligand, HLA-DR, on the tumor cells themselves. Finally, the cabozantinib/nivolumab combination was tested in comparison to cobimetinib/atezolizumab. Although both combinations showed tumor growth inhibition, cabozantinib/nivolumab had enhanced cytotoxic IFNγ and TNFα+ T cells. This pre-clinical in vivo data warrants testing the combination in clinical trials for patients with MSS-CRC., Competing Interests: Exelixis supplied cabozantinib and funded this study, with TP as Principal Investigator, but had no role in study design, data collection, data analysis, data interpretation, or writing of the report. WM is the principal investigator of a phase II clinical trial of cabozantinib in patients with refractory metastatic colorectal cancer that is sponsored by Exelixis NCT03542877. AL is the principal investigator of a phase II clinical trial of cabozantinib in combination with nivolumab in metastatic colorectal cancer which is co-sponsored by Bristol Myers Squibb and Exelixis NCT04963283. 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 © 2022 Lang, Leal, Marín-Jiménez, Hartman, Shulman, Navarro, Lewis, Capasso, Bagby, Yacob, MacBeth, Freed, Eckhardt, Jordan, Blatchford, Pelanda, Lieu, Messersmith and Pitts.)- Published
- 2022
- Full Text
- View/download PDF
19. ATM kinase inhibitor AZD0156 in combination with irinotecan and 5-fluorouracil in preclinical models of colorectal cancer.
- Author
-
Davis SL, Hartman SJ, Bagby SM, Schlaepfer M, Yacob BW, Tse T, Simmons DM, Diamond JR, Lieu CH, Leal AD, Cadogan EB, Hughes GD, Durant ST, Messersmith WA, and Pitts TM
- Subjects
- Humans, Irinotecan therapeutic use, Apoptosis, Cell Line, Tumor, G2 Phase Cell Cycle Checkpoints, Camptothecin, Ataxia Telangiectasia Mutated Proteins genetics, Fluorouracil pharmacology, Fluorouracil therapeutic use, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology
- Abstract
Background: AZD0156 is an oral inhibitor of ATM, a serine threonine kinase that plays a key role in DNA damage response (DDR) associated with double-strand breaks. Topoisomerase-I inhibitor irinotecan is used clinically to treat colorectal cancer (CRC), often in combination with 5-fluorouracil (5FU). AZD0156 in combination with irinotecan and 5FU was evaluated in preclinical models of CRC to determine whether low doses of AZD0156 enhance the cytotoxicity of irinotecan in chemotherapy regimens used in the clinic., Methods: Anti-proliferative effects of single-agent AZD0156, the active metabolite of irinotecan (SN38), and combination therapy were evaluated in 12 CRC cell lines. Additional assessment with clonogenic assay, cell cycle analysis, and immunoblotting were performed in 4 selected cell lines. Four colorectal cancer patient derived xenograft (PDX) models were treated with AZD0156, irinotecan, or 5FU alone and in combination for assessment of tumor growth inhibition (TGI). Immunofluorescence was performed on tumor tissues. The DDR mutation profile was compared across in vitro and in vivo models., Results: Enhanced effects on cellular proliferation and regrowth were observed with the combination of AZD0156 and SN38 in select models. In cell cycle analysis of these models, increased G2/M arrest was observed with combination treatment over either single agent. Immunoblotting results suggest an increase in DDR associated with irinotecan therapy, with a reduced effect noted when combined with AZD0156, which is more pronounced in some models. Increased TGI was observed with the combination of AZD0156 and irinotecan as compared to single-agent therapy in some PDX models. The DDR mutation profile was variable across models., Conclusions: AZD0156 and irinotecan provide a rational and active combination in preclinical colorectal cancer models. Variability across in vivo and in vitro results may be related to the variable DDR mutation profiles of the models evaluated. Further understanding of the implications of individual DDR mutation profiles may help better identify patients more likely to benefit from treatment with the combination of AZD0156 and irinotecan in the clinical setting., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
20. Preclinical Development of the Class-I-Selective Histone Deacetylase Inhibitor OKI-179 for the Treatment of Solid Tumors.
- Author
-
Diamond JR, Pitts TM, Ungermannova D, Nasveschuk CG, Zhang G, Phillips AJ, Bagby SM, Pafford J, Yacob BW, Newton TP, Tentler JJ, Gittleman B, Hartman SJ, DeMattei JA, Winkler JD, Wendt MK, Schiemann WP, Eckhardt SG, Liu X, and Piscopio AD
- Subjects
- Acetylation, Histone Deacetylase 1 metabolism, Histone Deacetylases metabolism, Histones metabolism, Humans, Histone Deacetylase Inhibitors pharmacology, Histone Deacetylase Inhibitors therapeutic use, Neoplasms drug therapy
- Abstract
Histone deacetylases (HDACs) play critical roles in epigenomic regulation, and histone acetylation is dysregulated in many human cancers. Although HDAC inhibitors are active in T-cell lymphomas, poor isoform selectivity, narrow therapeutic indices, and a deficiency of reliable biomarkers may contribute to the lack of efficacy in solid tumors. In this article, we report the discovery and preclinical development of the novel, orally bioavailable, class-I-selective HDAC inhibitor, OKI-179. OKI-179 and its cell active predecessor OKI-005 are thioester prodrugs of the active metabolite OKI-006, a unique congener of the natural product HDAC inhibitor largazole. OKI-006, OKI-005, and subsequently OKI-179, were developed through a lead candidate optimization program designed to enhance physiochemical properties without eroding potency and selectivity relative to largazole. OKI-005 displays antiproliferative activity in vitro with induction of apoptosis and increased histone acetylation, consistent with target engagement. OKI-179 showed antitumor activity in preclinical cancer models with a favorable pharmacokinetic profile and on-target pharmacodynamic effects. Based on its potency, desirable class I HDAC inhibition profile, oral bioavailability, and efficacy against a broad range of solid tumors, OKI-179 is currently being evaluated in a first-in-human phase I clinical trial with plans for continued clinical development in solid tumor and hematologic malignancies., (©2021 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
21. Modulating Enzyme Function via Dynamic Allostery within Biliverdin Reductase B.
- Author
-
Redzic JS, Duff MR, Blue A, Pitts TM, Agarwal P, and Eisenmesser EZ
- Abstract
The biliverdin reductase B (BLVRB) class of enzymes catalyze the NADPH-dependent reduction of multiple flavin substrates and are emerging as critical players in cellular redox regulation. However, the role of dynamics and allostery have not been addressed, prompting studies here that have revealed a position 15 Å away from the active site within human BLVRB (T164) that is inherently dynamic and can be mutated to control global micro-millisecond motions and function. By comparing the inherent dynamics through nuclear magnetic resonance (NMR) relaxation approaches of evolutionarily distinct BLVRB homologues and by applying our previously developed Relaxation And Single Site Multiple Mutations (RASSMM) approach that monitors both the functional and dynamic effects of multiple mutations to the single T164 site, we have discovered that the most dramatic mutagenic effects coincide with evolutionary changes and these modulate coenzyme binding. Thus, evolutionarily changing sites distal to the active site serve as dynamic "dials" to globally modulate motions and function. Despite the distal dynamic and functional coupling modulated by this site, micro-millisecond motions span an order of magnitude in their apparent kinetic rates of motions. Thus, global dynamics within BLVRB are a collection of partially coupled motions tied to catalytic function., Competing Interests: The 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 Redzic, Duff, Blue, Pitts, Agarwal and Eisenmesser.)
- Published
- 2021
- Full Text
- View/download PDF
22. Testing Cancer Immunotherapy in a Human Immune System Mouse Model: Correlating Treatment Responses to Human Chimerism, Therapeutic Variables and Immune Cell Phenotypes.
- Author
-
Marín-Jiménez JA, Capasso A, Lewis MS, Bagby SM, Hartman SJ, Shulman J, Navarro NM, Yu H, Rivard CJ, Wang X, Barkow JC, Geng D, Kar A, Yingst A, Tufa DM, Dolan JT, Blatchford PJ, Freed BM, Torres RM, Davila E, Slansky JE, Pelanda R, Eckhardt SG, Messersmith WA, Diamond JR, Lieu CH, Verneris MR, Wang JH, Kiseljak-Vassiliades K, Pitts TM, and Lang J
- Subjects
- Animals, Chimerism, Hematopoietic Stem Cell Transplantation, Humans, Lymphocyte Subsets immunology, Lymphocyte Subsets metabolism, Lymphoid Tissue immunology, Lymphoid Tissue metabolism, Mice, Mice, Inbred NOD, Mice, Knockout, Mice, SCID, Neoplasms etiology, Phenotype, Xenograft Model Antitumor Assays, Disease Models, Animal, Heterografts, Immune System, Immunotherapy adverse effects, Immunotherapy methods, Neoplasms immunology, Neoplasms therapy
- Abstract
Over the past decade, immunotherapies have revolutionized the treatment of cancer. Although the success of immunotherapy is remarkable, it is still limited to a subset of patients. More than 1500 clinical trials are currently ongoing with a goal of improving the efficacy of immunotherapy through co-administration of other agents. Preclinical, small-animal models are strongly desired to increase the pace of scientific discovery, while reducing the cost of combination drug testing in humans. Human immune system (HIS) mice are highly immune-deficient mouse recipients rtpeconstituted with human hematopoietic stem cells. These HIS-mice are capable of growing human tumor cell lines and patient-derived tumor xenografts. This model allows rapid testing of multiple, immune-related therapeutics for tumors originating from unique clinical samples. Using a cord blood-derived HIS-BALB/c-Rag2
null Il2rγnull SIRPαNOD (BRGS) mouse model, we summarize our experiments testing immune checkpoint blockade combinations in these mice bearing a variety of human tumors, including breast, colorectal, pancreatic, lung, adrenocortical, melanoma and hematological malignancies. We present in-depth characterization of the kinetics and subsets of the HIS in lymph and non-lymph organs and relate these to protocol development and immune-related treatment responses. Furthermore, we compare the phenotype of the HIS in lymph tissues and tumors. We show that the immunotype and amount of tumor infiltrating leukocytes are widely-variable and that this phenotype is tumor-dependent in the HIS-BRGS model. We further present flow cytometric analyses of immune cell subsets, activation state, cytokine production and inhibitory receptor expression in peripheral lymph organs and tumors. We show that responding tumors bear human infiltrating T cells with a more inflammatory signature compared to non-responding tumors, similar to reports of "responding" patients in human immunotherapy clinical trials. Collectively these data support the use of HIS mice as a preclinical model to test combination immunotherapies for human cancers, if careful attention is taken to both protocol details and data analysis., Competing Interests: The 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 Marín-Jiménez, Capasso, Lewis, Bagby, Hartman, Shulman, Navarro, Yu, Rivard, Wang, Barkow, Geng, Kar, Yingst, Tufa, Dolan, Blatchford, Freed, Torres, Davila, Slansky, Pelanda, Eckhardt, Messersmith, Diamond, Lieu, Verneris, Wang, Kiseljak-Vassiliades, Pitts and Lang.)- Published
- 2021
- Full Text
- View/download PDF
23. WEE1 Inhibition in Combination With Targeted Agents and Standard Chemotherapy in Preclinical Models of Pancreatic Ductal Adenocarcinoma.
- Author
-
Hartman SJ, Bagby SM, Yacob BW, Simmons DM, MacBeth M, Lieu CH, Davis SL, Leal AD, Tentler JJ, Diamond JR, Eckhardt SG, Messersmith WA, and Pitts TM
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal cancer with high incidences of p53 mutations. AZD1775 (adavosertib, previously MK-1775) is a small molecule WEE1 inhibitor that abrogates the G2M checkpoint and can potentially synergize with DNA damaging therapies commonly used in PDAC treatment. The purpose of this study was to identify combination partners for AZD1775, including standard chemotherapy or targeted agents, in PDAC preclinical models. Low powered preliminary screens demonstrated that two of the four PDX models responded better to the combinations of AZD1775 with irinotecan or capecitabine than to either single agent. Following the screens, two full powered PDAC PDX models of differing p53 status were tested with the combinations of AZD1775 and irinotecan or capecitabine. The combinations of AZD1775 and SN38 or 5-FU were also tested on PDAC cell lines. Cellular proliferation was measured using an IncuCyte Live Cell Imager and apoptosis was measured using a Caspase-Glo 3/7 assay. Flow cytometry was conducted to measure alterations in cell cycle distribution. Western blot analysis was used to determine the effects of the drug combinations on downstream effectors. In PDX models with mutated p53 status, there was significant tumor growth inhibition from the combination of AZD1775 with irinotecan or capecitabine ( P ≤ 0.03), while PDX models with wild type p53 did not show anti-tumor synergy from the same combinations ( P ≥ 0.08). The combination of AZD1775 with SN38 or 5-FU significantly decreased proliferation in all PDAC cell lines, and enhanced apoptosis in multiple cell lines. Cell cycle distribution was disrupted from the combination of AZD1775 with SN38 or 5-FU which was recorded as G2M arrest and decreased G1 phase. AZD1775 inhibited phospho-CDC2 and increased the expression of γ H2AX that was either maintained or enhanced after combination with SN38 or 5-FU. The combination of AZD1775 with irinotecan/SN38 or capecitabine/5-FU showed anti-tumor effects in vivo and in vitro in PDAC models. These results support further investigation for these combination strategies to enhance outcomes for PDAC patients., Competing Interests: The 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 Hartman, Bagby, Yacob, Simmons, MacBeth, Lieu, Davis, Leal, Tentler, Diamond, Eckhardt, Messersmith and Pitts.)
- Published
- 2021
- Full Text
- View/download PDF
24. RX-5902, a novel β-catenin modulator, potentiates the efficacy of immune checkpoint inhibitors in preclinical models of triple-negative breast Cancer.
- Author
-
Tentler JJ, Lang J, Capasso A, Kim DJ, Benaim E, Lee YB, Eisen A, Bagby SM, Hartman SJ, Yacob BW, Gittleman B, Pitts TM, Pelanda R, Eckhardt SG, and Diamond JR
- Subjects
- Animals, Apoptosis, Cell Proliferation, Drug Therapy, Combination, Female, Humans, Lymphocytes, Tumor-Infiltrating drug effects, Mice, Mice, Inbred BALB C, Mice, Nude, Triple Negative Breast Neoplasms metabolism, Triple Negative Breast Neoplasms pathology, Tumor Cells, Cultured, Tumor Microenvironment drug effects, Xenograft Model Antitumor Assays, beta Catenin metabolism, Gene Expression Regulation, Neoplastic drug effects, Immune Checkpoint Inhibitors pharmacology, Lymphocytes, Tumor-Infiltrating immunology, Piperazines pharmacology, Quinoxalines pharmacology, Triple Negative Breast Neoplasms drug therapy, Tumor Microenvironment immunology, beta Catenin antagonists & inhibitors
- Abstract
Background: Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype with limited systemic treatment options. RX-5902 is a novel anti-cancer agent that inhibits phosphorylated-p68 and thus attenuates nuclear β-catenin signaling. The purpose of this study was to evaluate the ability of β-catenin signaling blockade to enhance the efficacy of anti-CTLA-4 and anti-PD-1 immune checkpoint blockade in immunocompetent, preclinical models of TNBC., Methods: Treatment with RX-5902, anti-PD-1, anti-CTLA-4 or the combination was investigated in BALB/c mice injected with the 4 T1 TNBC cell line. Humanized BALB/c-Rag2
null Il2rγnull SIRPαNOD (hu-CB-BRGS) mice transplanted with a human immune system were implanted with MDA-MB-231 cells. Mice were randomized into treatment groups according to human hematopoietic chimerism and treated with RX-5902, anti-PD-1 or the combination. At sacrifice, bone marrow, lymph nodes, spleen and tumors were harvested for flow cytometry analysis of human immune cells., Results: The addition of RX-5902 to CTLA-4 or PD-1 inhibitors resulted in decreased tumor growth in the 4 T1 and human immune system and MDA-MB-231 xenograft models. Immunologic analyses demonstrated a significant increase in the number of activated T cells in tumor infiltrating lymphocytes (TILs) with RX-5902 treatment compared to vehicle (p < 0.05). In the RX-5902/nivolumab combination group, there was a significant increase in the percentage of CD4+ T cells in TILs and increased systemic granzyme B production (p < 0.01)., Conclusions: Conclusions: RX-5902 enhanced the efficacy of nivolumab in a humanized, preclinical model of TNBC. Several changes in immunologic profiles were noted in mice treated with RX-5902 and the combination, including an increase in activated TILs and a decrease in human myeloid populations, that are often associated with immunosuppression in a tumor microenvironment. RX-5902 also was shown to potentiate the effects of checkpoint inhibitors of CTLA4 and the PD-1 inhibitor in the 4 T-1 murine TNBC model. These findings indicate that RX-5902 may have important immunomodulatory, as well as anti-tumor activity, in TNBC when combined with a checkpoint inhibitor.- Published
- 2020
- Full Text
- View/download PDF
25. Bitter melon juice intake with gemcitabine intervention circumvents resistance to gemcitabine in pancreatic patient-derived xenograft tumors.
- Author
-
Dhar D, Raina K, Kumar D, Wempe MF, Bagby SM, Pitts TM, Orlicky DJ, Agarwal C, Messersmith WA, and Agarwal R
- Subjects
- Animals, Antimetabolites, Antineoplastic pharmacology, Apoptosis, Carcinoma, Pancreatic Ductal pathology, Cell Proliferation, Deoxycytidine pharmacology, Female, Humans, Mice, Mice, Nude, Neoplasm Invasiveness, Pancreatic Neoplasms pathology, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Gemcitabine, Carcinoma, Pancreatic Ductal drug therapy, Deoxycytidine analogs & derivatives, Disease Models, Animal, Drug Resistance, Neoplasm drug effects, Momordica charantia chemistry, Pancreatic Neoplasms drug therapy, Plant Extracts pharmacology
- Abstract
Chemoresistance to gemcitabine (GEM)-a frontline chemotherapeutic, resulting from its dysfunctional uptake and metabolism in cancer cells, is a major contributing factor for failed therapy in pancreatic cancer (PanC) patients. Therefore, there is an urgent need for agents that could reverse GEM resistance and allow continued chemosensitivity to the drug. We employed natural nontoxic agent (with anti-PanC potential) bitter melon juice (BMJ) and GEM to examine their combinatorial benefits against tumorigenesis of PanC patient-derived xenograft (PDX)-pancreatic ductal adenocarcinomas explants PDX272 (wild-type KRAS), PDX271 (mutant KRAS and SMAD4), and PDX266 (mutant KRAS). Anti-PanC efficacy of single agents vs combination in the three tumor explants, both at the end of active dosing regimen and following a drug-washout phase were compared. In animal studies, GEM alone treatment significantly inhibited PDX tumor growth, but effects were not sustained, as GEM-treated tumors exhibited regrowth posttreatment termination. However, combination-regimen displayed enhanced and sustained efficacy. Mechanistic assessments revealed that overcoming GEM resistance by coadministration with BMJ was possibly due to modulation of GEM transport/metabolism pathway molecules (ribonucleotide reductase regulatory subunit M1, human equilibrative nucleoside transporter 1, and deoxycytidine kinase). Study outcomes, highlighting significantly higher and sustained efficacy of GEM in combination with BMJ, make a compelling case for a clinical trial in PanC patients, wherein BMJ could be combined with GEM to target and overcome GEM resistance. In addition, given their specific effectiveness against KRAS-mutant tumors, this combination could be potentially beneficial to a broader PanC patient population., (© 2020 Wiley Periodicals LLC.)
- Published
- 2020
- Full Text
- View/download PDF
26. Preclinical and Dose-Finding Phase I Trial Results of Combined Treatment with a TORC1/2 Inhibitor (TAK-228) and Aurora A Kinase Inhibitor (Alisertib) in Solid Tumors.
- Author
-
Davis SL, Ionkina AA, Bagby SM, Orth JD, Gittleman B, Marcus JM, Lam ET, Corr BR, O'Bryant CL, Glode AE, Tan AC, Kim J, Tentler JJ, Capasso A, Lopez KL, Gustafson DL, Messersmith WA, Leong S, Eckhardt SG, Pitts TM, and Diamond JR
- Subjects
- Aged, Animals, Antineoplastic Combined Chemotherapy Protocols adverse effects, Aurora Kinase A antagonists & inhibitors, Aurora Kinase A metabolism, Azepines adverse effects, Benzoxazoles adverse effects, Cell Line, Tumor, Drug Resistance, Neoplasm drug effects, Female, Humans, Male, Maximum Tolerated Dose, Mechanistic Target of Rapamycin Complex 1 antagonists & inhibitors, Mechanistic Target of Rapamycin Complex 1 metabolism, Mechanistic Target of Rapamycin Complex 2 antagonists & inhibitors, Mechanistic Target of Rapamycin Complex 2 metabolism, Mice, Middle Aged, Neoplasms pathology, Protein Kinase Inhibitors adverse effects, Pyrimidines adverse effects, Xenograft Model Antitumor Assays, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Azepines administration & dosage, Benzoxazoles administration & dosage, Neoplasms drug therapy, Protein Kinase Inhibitors administration & dosage, Pyrimidines administration & dosage
- Abstract
Purpose: The purpose of this study was to evaluate the rational combination of TORC1/2 inhibitor TAK-228 and Aurora A kinase inhibitor alisertib in preclinical models of triple-negative breast cancer (TNBC) and to conduct a phase I dose escalation trial in patients with advanced solid tumors., Experimental Design: TNBC cell lines and patient-derived xenograft (PDX) models were treated with alisertib, TAK-228, or the combination and evaluated for changes in proliferation, cell cycle, mTOR pathway modulation, and terminal cellular fate, including apoptosis and senescence. A phase I clinical trial was conducted in patients with advanced solid tumors treated with escalating doses of alisertib and TAK-228 using a 3+3 design to determine the maximum tolerated dose (MTD)., Results: The combination of TAK-228 and alisertib resulted in decreased proliferation and cell-cycle arrest in TNBC cell lines. Treatment of TNBC PDX models resulted in significant tumor growth inhibition and increased apoptosis with the combination. In the phase I dose escalation study, 18 patients with refractory solid tumors were enrolled. The MTD was alisertib 30 mg b.i.d. days 1 to 7 of a 21-day cycle and TAK-228 2 mg daily, continuous dosing. The most common treatment-related adverse events were neutropenia, fatigue, nausea, rash, mucositis, and alopecia., Conclusions: The addition of TAK-228 to alisertib potentiates the antitumor activity of alisertib in vivo , resulting in increased cell death and apoptosis. The combination is tolerable in patients with advanced solid tumors and should be evaluated further in expansion cohorts with additional pharmacodynamic assessment., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
27. Structure, dynamics and function of the evolutionarily changing biliverdin reductase B family.
- Author
-
Duff MR, Redzic JS, Ryan LP, Paukovich N, Zhao R, Nix JC, Pitts TM, Agarwal P, and Eisenmesser EZ
- Subjects
- Crystallography, X-Ray, Humans, Models, Molecular, Oxidoreductases Acting on CH-CH Group Donors isolation & purification, Protein Conformation, Oxidoreductases Acting on CH-CH Group Donors chemistry, Oxidoreductases Acting on CH-CH Group Donors metabolism, Thermodynamics
- Abstract
Biliverdin reductase B (BLVRB) family members are general flavin reductases critical in maintaining cellular redox with recent findings revealing that BLVRB alone can dictate cellular fate. However, as opposed to most enzymes, the BLVRB family remains enigmatic with an evolutionarily changing active site and unknown structural and functional consequences. Here, we applied a multi-faceted approach that combines X-ray crystallography, NMR and kinetics methods to elucidate the structural and functional basis of the evolutionarily changing BLVRB active site. Using a panel of three BLVRB isoforms (human, lemur and hyrax) and multiple human BLVRB mutants, our studies reveal a novel evolutionary mechanism where coenzyme 'clamps' formed by arginine side chains at two co-evolving positions within the active site serve to slow coenzyme release (Positions 14 and 78). We find that coenzyme release is further slowed by the weaker binding substrate, resulting in relatively slow turnover numbers. However, different BLVRB active sites imposed by either evolution or mutagenesis exhibit a surprising inverse relationship between coenzyme release and substrate turnover that is independent of the faster chemical step of hydride transfer also measured here. Collectively, our studies have elucidated the role of the evolutionarily changing BLVRB active site that serves to modulate coenzyme release and has revealed that coenzyme release is coupled to substrate turnover., (© The Author(s) 2020. Published by Oxford University Press on behalf of the Japanese Biochemical Society. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
28. First-in-Class Inhibitors of Oncogenic CHD1L with Preclinical Activity against Colorectal Cancer.
- Author
-
Abbott JM, Zhou Q, Esquer H, Pike L, Broneske TP, Rinaldetti S, Abraham AD, Ramirez DA, Lunghofer PJ, Pitts TM, Regan DP, Tan AC, Gustafson DL, Messersmith WA, and LaBarbera DV
- Subjects
- Adenocarcinoma mortality, Animals, Antineoplastic Agents pharmacology, Antineoplastic Agents toxicity, Apoptosis, Cell Line, Tumor, Colorectal Neoplasms mortality, DNA Damage, DNA Helicases genetics, DNA Helicases physiology, DNA-Binding Proteins genetics, DNA-Binding Proteins physiology, Drug Screening Assays, Antitumor, Enzyme Inhibitors pharmacology, Enzyme Inhibitors toxicity, Epithelial-Mesenchymal Transition drug effects, Epithelial-Mesenchymal Transition physiology, Female, Gene Expression Regulation, Neoplastic drug effects, Gene Knockdown Techniques, High-Throughput Screening Assays, Humans, Kaplan-Meier Estimate, Mice, Neoplasm Proteins genetics, Neoplasm Proteins physiology, Organoids drug effects, RNA, Messenger biosynthesis, RNA, Neoplasm biosynthesis, Recombinant Proteins metabolism, Small Molecule Libraries, TCF Transcription Factors physiology, Transcription, Genetic drug effects, Wnt Signaling Pathway drug effects, Wnt Signaling Pathway genetics, Wnt Signaling Pathway physiology, Adenocarcinoma drug therapy, Antineoplastic Agents therapeutic use, Colorectal Neoplasms drug therapy, DNA Helicases antagonists & inhibitors, DNA-Binding Proteins antagonists & inhibitors, Enzyme Inhibitors therapeutic use, Neoplasm Proteins antagonists & inhibitors
- Abstract
Since the discovery of CHD1L in 2008, it has emerged as an oncogene implicated in the pathology and poor prognosis of a variety of cancers, including gastrointestinal cancers. However, a mechanistic understanding of CHD1L as a driver of colorectal cancer has been limited. Until now, there have been no reported inhibitors of CHD1L, also limiting its development as a molecular target. We sought to characterize the clinicopathologic link between CHD1L and colorectal cancer, determine the mechanism(s) by which CHD1L drives malignant colorectal cancer, and discover the first inhibitors with potential for novel treatments for colorectal cancer. The clinicopathologic characteristics associated with CHD1L expression were evaluated using microarray data from 585 patients with colorectal cancer. Further analysis of microarray data indicated that CHD1L may function through the Wnt/TCF pathway. Thus, we conducted knockdown and overexpression studies with CHD1L to determine its role in Wnt/TCF-driven epithelial-to-mesenchymal transition (EMT). We performed high-throughput screening (HTS) to identify the first CHD1L inhibitors. The mechanism of action, antitumor efficacy, and drug-like properties of lead CHD1L inhibitors were determined using biochemical assays, cell models, tumor organoids, patient-derived tumor organoids, and in vivo pharmacokinetics and pharmacodynamics. Lead CHD1L inhibitors display potent in vitro antitumor activity by reversing TCF-driven EMT. The best lead CHD1L inhibitor possesses drug-like properties in pharmacokinetic/pharmacodynamic mouse models. This work validates CHD1L as a druggable target and establishes a novel therapeutic strategy for the treatment of colorectal cancer., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
29. Wee1 Inhibition Enhances the Anti-Tumor Effects of Capecitabine in Preclinical Models of Triple-Negative Breast Cancer.
- Author
-
Pitts TM, Simmons DM, Bagby SM, Hartman SJ, Yacob BW, Gittleman B, Tentler JJ, Cittelly D, Ormond DR, Messersmith WA, Eckhardt SG, and Diamond JR
- Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype defined by lack of hormone receptor expression and non-amplified HER2. Adavosertib (AZD1775) is a potent, small-molecule, ATP-competitive inhibitor of the Wee1 kinase that potentiates the activity of many DNA-damaging chemotherapeutics and is currently in clinical development for multiple indications. The purpose of this study was to investigate the combination of AZD1775 and capecitabine/5FU in preclinical TNBC models. TNBC cell lines were treated with AZD1775 and 5FU and cellular proliferation was assessed in real-time using IncuCyte
® Live Cell Analysis. Apoptosis was assessed via the Caspase-Glo 3/7 assay system. Western blotting was used to assess changes in expression of downstream effectors. TNBC patient-derived xenograft (PDX) models were treated with AZD1775, capecitabine, or the combination and assessed for tumor growth inhibition. From the initial PDX screen, two of the four TNBC PDX models demonstrated a better response in the combination treatment than either of the single agents. As confirmation, two PDX models were expanded for statistical comparison. Both PDX models demonstrated a significant growth inhibition in the combination versus either of the single agents. (TNBC012, p < 0.05 combo vs. adavosertib or capecitabine, TNBC013, p < 0.01 combo vs. adavosertib or capecitabine.) An enhanced anti-proliferative effect was observed in the adavosertib/5FU combination treatment as measured by live cell analysis. An increase in apoptosis was observed in two of the four cell lines in the combination when compared to single-agent treatment. Treatment with adavosertib as a single agent resulted in a decrease in p-CDC2 in a dose-dependent manner that was also observed in the combination treatment. An increase in γH2AX in two of the four cell lines tested was also observed. No significant changes were observed in Bcl-xL following treatment in any of the cell lines. The combination of adavosertib and capecitabine/5FU demonstrated enhanced combination effects both in vitro and in vivo in preclinical models of TNBC. These results support the clinical investigation of this combination in patients with TNBC, including those with brain metastasis given the CNS penetration of both agents.- Published
- 2020
- Full Text
- View/download PDF
30. Development of an Adrenocortical Cancer Humanized Mouse Model to Characterize Anti-PD1 Effects on Tumor Microenvironment.
- Author
-
Lang J, Capasso A, Jordan KR, French JD, Kar A, Bagby SM, Barbee J, Yacob BW, Head LS, Tompkins KD, Freed BM, Somerset H, Clark TJ, Pitts TM, Messersmith WA, Eckhardt SG, Wierman ME, Leong S, and Kiseljak-Vassiliades K
- Subjects
- Adrenal Cortex Neoplasms immunology, Adrenal Cortex Neoplasms pathology, Adrenocortical Carcinoma immunology, Adrenocortical Carcinoma pathology, Animals, Antineoplastic Agents, Immunological pharmacology, Apoptosis, Cell Proliferation, Female, Humans, Immunotherapy, Lymphocytes, Tumor-Infiltrating drug effects, Lymphocytes, Tumor-Infiltrating immunology, Mice, Mice, Inbred BALB C, Mice, Nude, Programmed Cell Death 1 Receptor immunology, Tumor Cells, Cultured, Tumor Microenvironment drug effects, Xenograft Model Antitumor Assays, Adrenal Cortex Neoplasms drug therapy, Adrenocortical Carcinoma drug therapy, Antibodies, Monoclonal, Humanized pharmacology, Disease Models, Animal, Programmed Cell Death 1 Receptor antagonists & inhibitors, Tumor Microenvironment immunology
- Abstract
Context: Although the development of immune checkpoint inhibitors has transformed treatment strategies of several human malignancies, research models to study immunotherapy in adrenocortical carcinoma (ACC) are lacking., Objective: To explore the effect of anti-PD1 immunotherapy on the alteration of the immune milieu in ACC in a newly generated preclinical model and correlate with the response of the matched patient., Design, Setting, and Intervention: To characterize the CU-ACC2-M2B patient-derived xenograft in a humanized mouse model, evaluate the effect of a PD-1 inhibitor therapy, and compare it with the CU-ACC2 patient with metastatic disease., Results: Characterization of the CU-ACC2-humanized cord blood-BALB/c-Rag2nullIl2rγnullSirpaNOD model confirmed ACC origin and match with the original human tumor. Treatment of the mice with pembrolizumab demonstrated significant tumor growth inhibition (60%) compared with controls, which correlated with increased tumor infiltrating lymphocyte activity, with an increase of human CD8+ T cells (P < 0.05), HLA-DR+ T cells (P < 0.05) as well as Granzyme B+ CD8+ T cells (<0.001). In parallel, treatment of the CU-ACC2 patient, who had progressive disease, demonstrated a partial response with 79% to 100% reduction in the size of target lesions, and no new sites of metastasis. Pretreatment analysis of the patient's metastatic liver lesion demonstrated abundant intratumoral CD8+ T cells by immunohistochemistry., Conclusions: Our study reports the first humanized ACC patient-derived xenograft mouse model, which may be useful to define mechanisms and biomarkers of response and resistance to immune-based therapies, to ultimately provide more personalized care for patients with ACC., (© Endocrine Society 2019. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
31. First-in-Class Phosphorylated-p68 Inhibitor RX-5902 Inhibits β-Catenin Signaling and Demonstrates Antitumor Activity in Triple-Negative Breast Cancer.
- Author
-
Capasso A, Bagby SM, Dailey KL, Currimjee N, Yacob BW, Ionkina A, Frank JG, Kim DJ, George C, Lee YB, Benaim E, Gittleman B, Hartman SJ, Tan AC, Kim J, Pitts TM, Eckhardt SG, Tentler JJ, and Diamond JR
- Subjects
- Animals, Antineoplastic Agents pharmacology, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, Mice, Myeloid Cell Leukemia Sequence 1 Protein metabolism, Phosphorylation, Piperazines pharmacology, Quinoxalines pharmacology, Triple Negative Breast Neoplasms metabolism, Xenograft Model Antitumor Assays, beta Catenin metabolism, Antineoplastic Agents administration & dosage, Piperazines administration & dosage, Quinoxalines administration & dosage, Triple Negative Breast Neoplasms drug therapy, Wnt Signaling Pathway drug effects, eIF-2 Kinase metabolism
- Abstract
RX-5902 is a first-in-class anticancer agent targeting phosphorylated-p68 and attenuating nuclear shuttling of β-catenin. The purpose of this study was to evaluate the efficacy of RX-5902 in preclinical models of triple-negative breast cancer (TNBC) and to explore effects on β-catenin expression. A panel of 18 TNBC cell lines was exposed to RX-5902, and changes in proliferation, apoptosis, cellular ploidy, and effector protein expression were assessed. Gene expression profiling was used in sensitive and resistant cell lines with pathway analysis to explore pathways associated with sensitivity to RX-5902. The activity of RX-5902 was confirmed in vivo in cell line and patient-derived tumor xenograft (PDX) models. RX-5902 demonstrated potent antiproliferative activity in vitro against TNBC cell lines with an average IC
50 of 56 nmol/L in sensitive cell lines. RX-5902 treatment resulted in the induction of apoptosis, G2 -M cell-cycle arrest, and aneuploidy in a subset of cell lines. RX-5902 was active in vivo against TNBC PDX models, and treatment resulted in a decrease in nuclear β-catenin. RX-5902 exhibited dose-proportional pharmacokinetics and plasma and tumor tissue in nude mice. Pathway analysis demonstrated an increase in the epithelial-to-mesenchymal transformation (EMT), TGFβ, and Wnt/β-catenin pathways associated with sensitivity to RX-5902. RX-5902 is active against in vitro and in vivo preclinical models of TNBC. Target engagement was confirmed with decreases in nuclear β-catenin and MCL-1 observed, confirming the proposed mechanism of action. This study supports the continued investigation of RX-5902 in TNBC and combinations with immunotherapy., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
32. Targeting PDZ-binding kinase is anti-tumorigenic in novel preclinical models of ACC.
- Author
-
Kar A, Zhang Y, Yacob BW, Saeed J, Tompkins KD, Bagby SM, Pitts TM, Somerset H, Leong S, Wierman ME, and Kiseljak-Vassiliades K
- Subjects
- Adrenocortical Carcinoma genetics, Animals, Antineoplastic Agents pharmacology, Apoptosis, Carcinogenesis, Cell Cycle Checkpoints, Cell Line, Tumor, Forkhead Box Protein M1 antagonists & inhibitors, Forkhead Box Protein M1 metabolism, Gene Expression Regulation, Neoplastic, Humans, Indolizines pharmacology, Indolizines therapeutic use, Mice, Mitogen-Activated Protein Kinase Kinases genetics, Mitogen-Activated Protein Kinase Kinases metabolism, Neoplasms, Experimental, Phosphorylation, Prognosis, Quinoxalines pharmacology, Quinoxalines therapeutic use, RNA, Small Interfering genetics, Signal Transduction, Xenograft Model Antitumor Assays, Adrenocortical Carcinoma drug therapy, Adrenocortical Carcinoma pathology, Antineoplastic Agents therapeutic use, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors
- Abstract
Adrenocortical carcinoma (ACC) is an aggressive orphan malignancy with less than 35% 5-year survival and 75% recurrence. Surgery remains the primary therapy and mitotane, an adrenolytic, is the only FDA-approved drug with wide-range toxicities and poor tolerability. There are no targeted agents available to date. For the last three decades, H295R cell line and its xenograft were the only available preclinical models. We recently developed two new ACC patient-derived xenograft mouse models and corresponding cell lines (CU-ACC1 and CU-ACC2) to advance research in the field. Here, we have utilized these novel models along with H295R cells to establish the mitotic PDZ-binding kinase (PBK) as a promising therapeutic target. PBK is overexpressed in ACC samples and correlates with poor survival. We show that PBK is regulated by FOXM1 and targeting PBK via shRNA decreased cell proliferation, clonogenicity and anchorage-independent growth in ACC cell lines. PBK silencing inhibited pAkt, pp38MAPK and pHistone H3 altering the cell cycle. Therapeutically, targeting PBK with the small-molecule inhibitor HITOPK032 phenocopied PBK-specific modulation of pAkt and pHistone H3, but also induced apoptosis via activation of JNK. Consistent with in vitro findings, treatment of CU-ACC1 PDXs with HITOPK032 significantly reduced tumor growth by 5-fold (P < 0.01). Treated tumor tissues demonstrated increased rates of apoptosis and JNK activation, with decreased pAkt and Histone H3 phosphorylation, consistent with effects observed in ACC cell lines. Together these studies elucidate the mechanism of PBK in ACC tumorigenesis and establish the potential therapeutic potential of HITOPK032 in ACC patients.
- Published
- 2019
- Full Text
- View/download PDF
33. Pancreatic Tumor Microenvironment Modulation by EphB4-ephrinB2 Inhibition and Radiation Combination.
- Author
-
Lennon S, Oweida A, Milner D, Phan AV, Bhatia S, Van Court B, Darragh L, Mueller AC, Raben D, Martínez-Torrecuadrada JL, Pitts TM, Somerset H, Jordan KR, Hansen KC, Williams J, Messersmith WA, Schulick RD, Owens P, Goodman KA, and Karam SD
- Subjects
- Animals, Antineoplastic Agents, Immunological pharmacology, Antineoplastic Agents, Immunological therapeutic use, Biomarkers, Cell Line, Tumor, Disease Models, Animal, Ephrin-B2 antagonists & inhibitors, Ephrin-B2 genetics, Female, Flow Cytometry, Gene Expression, Gene Knockdown Techniques, Humans, Immunohistochemistry, Mice, Molecular Targeted Therapy adverse effects, Molecular Targeted Therapy methods, Neutrophils immunology, Neutrophils metabolism, Pancreatic Neoplasms therapy, Radiotherapy adverse effects, Radiotherapy methods, Receptor, EphB4 antagonists & inhibitors, Receptor, EphB4 genetics, Signal Transduction drug effects, T-Lymphocytes, Regulatory immunology, T-Lymphocytes, Regulatory metabolism, Xenograft Model Antitumor Assays, Ephrin-B2 metabolism, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms pathology, Receptor, EphB4 metabolism, Tumor Microenvironment drug effects, Tumor Microenvironment radiation effects
- Abstract
Purpose: A driving factor in pancreatic ductal adenocarcinoma (PDAC) treatment resistance is the tumor microenvironment, which is highly immunosuppressive. One potent immunologic adjuvant is radiotherapy. Radiation, however, has also been shown to induce immunosuppressive factors, which can contribute to tumor progression and formation of fibrotic tumor stroma. To capitalize on the immunogenic effects of radiation and obtain a durable tumor response, radiation must be rationally combined with targeted therapies to mitigate the influx of immunosuppressive cells and fibrosis. One such target is ephrinB2, which is overexpressed in PDAC and correlates negatively with prognosis. Experimental Design: On the basis of previous studies of ephrinB2 ligand-EphB4 receptor signaling, we hypothesized that inhibition of ephrinB2-EphB4 combined with radiation can regulate the microenvironment response postradiation, leading to increased tumor control in PDAC. This hypothesis was explored using both cell lines and in vivo human and mouse tumor models., Results: Our data show this treatment regimen significantly reduces regulatory T-cell, macrophage, and neutrophil infiltration and stromal fibrosis, enhances effector T-cell activation, and decreases tumor growth. Furthermore, our data show that depletion of regulatory T cells in combination with radiation reduces tumor growth and fibrosis., Conclusions: These are the first findings to suggest that in PDAC, ephrinB2-EphB4 interaction has a profibrotic, protumorigenic role, presenting a novel and promising therapeutic target., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
34. Characterization of immune responses to anti-PD-1 mono and combination immunotherapy in hematopoietic humanized mice implanted with tumor xenografts.
- Author
-
Capasso A, Lang J, Pitts TM, Jordan KR, Lieu CH, Davis SL, Diamond JR, Kopetz S, Barbee J, Peterson J, Freed BM, Yacob BW, Bagby SM, Messersmith WA, Slansky JE, Pelanda R, and Eckhardt SG
- Subjects
- Animals, Antineoplastic Agents, Immunological pharmacology, CD8-Positive T-Lymphocytes immunology, Cell Line, Tumor, Colorectal Neoplasms immunology, Female, Humans, Lymphocytes, Tumor-Infiltrating immunology, Mice, Nude, Nivolumab pharmacology, Triple Negative Breast Neoplasms immunology, Tumor Microenvironment immunology, Xenograft Model Antitumor Assays, Antineoplastic Agents, Immunological therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Colorectal Neoplasms drug therapy, Disease Models, Animal, Histone Deacetylase Inhibitors therapeutic use, Nivolumab therapeutic use, Programmed Cell Death 1 Receptor antagonists & inhibitors, Triple Negative Breast Neoplasms drug therapy
- Abstract
Background: The success of agents that reverse T-cell inhibitory signals, such as anti-PD-1/PD-L1 therapies, has reinvigorated cancer immunotherapy research. However, since only a minority of patients respond to single-agent therapies, methods to test the potential anti-tumor activity of rational combination therapies are still needed. Conventional murine xenograft models have been hampered by their immune-compromised status; thus, we developed a hematopoietic humanized mouse model, hu-CB-BRGS, and used it to study anti-tumor human immune responses to triple-negative breast cancer (TNBC) cell line and patient-derived colorectal cancer (CRC) xenografts (PDX)., Methods: BALB/c-Rag2
null Il2rγnull SIRPαNOD (BRGS) pups were humanized through transplantation of cord blood (CB)-derived CD34+ cells. Mice were evaluated for human chimerism in the blood and assigned into experimental untreated or nivolumab groups based on chimerism. TNBC cell lines or tumor tissue from established CRC PDX models were implanted into both flanks of humanized mice and treatments ensued once tumors reached a volume of ~150mm3 . Tumors were measured twice weekly. At end of study, immune organs and tumors were collected for immunological assessment., Results: Humanized PDX models were successfully established with a high frequency of tumor engraftment. Humanized mice treated with anti-PD-1 exhibited increased anti-tumor human T-cell responses coupled with decreased Treg and myeloid populations that correlated with tumor growth inhibition. Combination therapies with anti-PD-1 treatment in TNBC-bearing mice reduced tumor growth in multi-drug cohorts. Finally, as observed in human colorectal patients, anti-PD-1 therapy had a strong response to a microsatellite-high CRC PDX that correlated with a higher number of human CD8+ IFNγ+ T cells in the tumor., Conclusion: Hu-CB-BRGS mice represent an in vivo model to study immune checkpoint blockade to human tumors. The human immune system in the mice is inherently suppressed, similar to a tumor microenvironment, and thus allows growth of human tumors. However, the suppression can be released by anti-PD-1 therapies and inhibit tumor growth of some tumors. The model offers ample access to lymph and tumor cells for in-depth immunological analysis. The tumor growth inhibition correlates with increased CD8 IFNγ+ tumor infiltrating T cells. These hu-CB-BRGS mice provide a relevant preclinical animal model to facilitate prioritization of hypothesis-driven combination immunotherapies.- Published
- 2019
- Full Text
- View/download PDF
35. A Phase I Dose-Escalation Study of Linsitinib (OSI-906), a Small-Molecule Dual Insulin-Like Growth Factor-1 Receptor/Insulin Receptor Kinase Inhibitor, in Combination with Irinotecan in Patients with Advanced Cancer.
- Author
-
Davis SL, Eckhardt SG, Diamond JR, Messersmith WA, Dasari A, Weekes CD, Lieu CH, Kane M, Choon Tan A, Pitts TM, and Leong S
- Subjects
- Adult, Aged, Female, Humans, Imidazoles pharmacology, Irinotecan pharmacology, Male, Middle Aged, Pyrazines pharmacology, Imidazoles therapeutic use, Insulin-Like Growth Factor I drug effects, Irinotecan therapeutic use, Pyrazines therapeutic use
- Abstract
Lessons Learned: The maximum tolerated dose of the combination of linsitinib and irinotecan is linsitinib 450 mg daily on days 1-3 every 7 days and irinotecan 125 mg/m
2 days 1 and 8 of a 21-day cycle.The adverse effects associated with the combination are not significantly increased beyond what is expected of each drug as a single agent.Multiple negative trials of insulin-like growth factor-1 receptor inhibitors performed in unselected patient populations led to the early discontinuation of linistinib development and this trial.Earlier integration of assessment of potential predictive biomarkers into clinical trials, as was planned in this study, is vital to the development of targeted therapies in oncology., Background: This phase I dose-escalation study was designed to evaluate the safety and tolerability of the combination of irinotecan and insulin-like growth factor-1 receptor (IGF-1R) inhibitor linsitinib in patients with advanced cancer refractory to standard therapy., Methods: Dose escalation in three specified dose levels was performed according to a standard 3 + 3 design. Dose levels were as follows: (a) linsitinib 400 mg and irinotecan 100 mg/m2 , (b) linsitinib 450 mg and irinotecan 100 mg/m2 , and (c) linsitinib 450 mg and irinotecan 125 mg/m2 . Linisitinib was administered once daily on days 1-3, 8-10, and 15-17, and irinotecan on days 1 and 8. Assessment of a candidate predictive biomarker was planned in all patients, with further evaluation in an expansion cohort of advanced colorectal cancer., Results: A total of 17 patients were treated, with 1 patient in both cohort 2 and 3 experiencing dose-limiting toxicity. Linsitinib 450 mg and irinotecan 125 mg/m2 was the maximum tolerated dose. Sixteen (94%) patients experienced at least one treatment-related adverse event. Neutropenia was the only grade >3 toxicity (4%). No significant hyperglycemia or QT interval prolongation was noted. No objective responses were observed; 47% ( n = 8) had stable disease with median duration of 5.25 months., Conclusion: Although the combination was determined safe, the study was halted due to termination of linsitinib development, and biomarker testing was not performed., (© AlphaMed Press; the data published online to support this summary are the property of the authors.)- Published
- 2018
- Full Text
- View/download PDF
36. Dual compartmental targeting of cell cycle and angiogenic kinases in colorectal cancer models.
- Author
-
Capasso A, Pitts TM, Klauck PJ, Bagby SM, Westbrook L, Kaplan J, Soleimani M, Spreafico A, Tentler JJ, Diamond JR, Arcaroli JJ, Messersmith WA, Eckhardt SG, and Leong S
- Subjects
- Angiogenesis Inhibitors pharmacology, Animals, Apoptosis drug effects, Cell Cycle drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Colorectal Neoplasms enzymology, Female, G2 Phase Cell Cycle Checkpoints drug effects, Humans, M Phase Cell Cycle Checkpoints drug effects, Mice, Mice, Nude, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Colorectal Neoplasms drug therapy, Protein Kinase Inhibitors pharmacology, Pyrazoles pharmacology, Pyrimidines pharmacology
- Abstract
Cancer is a disease caused by several factors characterized by uncontrolled cell division, growth, and survival. ENMD-2076, is a novel orally active small molecule multikinase inhibitor targeting angiogenesis, proliferation, and the cell cycle. It is selectively active against the mitotic kinases aurora A and B, and kinases responsible for angiogenesis including VEGFR2/KDR and FGFR1 and 2. ENMD-2076 has been shown to inhibit tumor growth and prevent angiogenesis in vitro and in vivo in preclinical cancer models. Moreover, in a phase I trial, ENMD-2076 was well tolerated, exhibited a linear pharmacokinetic profile, and showed a promising antitumor activity in a number of solid tumors. In this study, we show that ENMD-2076 has antiproliferative effects, causes cell cycle arrest, and has activity in preclinical models of colorectal cancer (CRC), including patient-derived xenograft (PDX) models. Forty-seven human CRC cell lines were exposed in vitro to ENMD-2076 and analyzed for effects on cell cycle, apoptosis, and downstream effector proteins. The drug was then tested against 20 human CRC PDX models to further evaluate in-vivo antitumor activity. We show that ENMD-2076 exhibits a broad range of activity against a large panel of CRC cell lines with varying molecular characteristics. Mechanistically, ENMD-2076 exposure resulted in a G2/M cell cycle arrest, an increase in aneuploidy, and cell death in responsive cell lines. In addition, ENMD-2076 treatment resulted in a promising antitumor activity in CRC PDX models. These results support the continued development of ENMD-2076 in CRC including further exploration of rational combinations.
- Published
- 2018
- Full Text
- View/download PDF
37. Cabozantinib Exhibits Potent Antitumor Activity in Colorectal Cancer Patient-Derived Tumor Xenograft Models via Autophagy and Signaling Mechanisms.
- Author
-
Scott AJ, Arcaroli JJ, Bagby SM, Yahn R, Huber KM, Serkova NJ, Nguyen A, Kim J, Thorburn A, Vogel J, Quackenbush KS, Capasso A, Schreiber A, Blatchford P, Klauck PJ, Pitts TM, Eckhardt SG, and Messersmith WA
- Subjects
- Animals, Apoptosis, Biomarkers, Tumor, Cell Line, Tumor, Colorectal Neoplasms diagnostic imaging, Colorectal Neoplasms drug therapy, Disease Models, Animal, Female, Humans, Magnetic Resonance Imaging, Mice, Neovascularization, Pathologic drug therapy, Neovascularization, Pathologic metabolism, Phosphatidylinositol 3-Kinases metabolism, Positron-Emission Tomography, Proto-Oncogene Proteins c-akt metabolism, TOR Serine-Threonine Kinases, Xenograft Model Antitumor Assays, Anilides pharmacology, Antineoplastic Agents pharmacology, Autophagy drug effects, Colorectal Neoplasms metabolism, Colorectal Neoplasms pathology, Protein Kinase Inhibitors pharmacology, Pyridines pharmacology, Signal Transduction drug effects
- Abstract
Antiangiogenic therapy used in treatment of metastatic colorectal cancer (mCRC) inevitably succumbs to treatment resistance. Upregulation of MET may play an essential role to acquired anti-VEGF resistance. We previously reported that cabozantinib (XL184), an inhibitor of receptor tyrosine kinases (RTK) including MET, AXL, and VEGFR2, had potent antitumor effects in mCRC patient-derived tumor explant models. In this study, we examined the mechanisms of cabozantinib sensitivity, using regorafenib as a control. The tumor growth inhibition index (TGII) was used to compare treatment effects of cabozantinib 30 mg/kg daily versus regorafenib 10 mg/kg daily for a maximum of 28 days in 10 PDX mouse models. In vivo angiogenesis and glucose uptake were assessed using dynamic contrast-enhanced (DCE)-MRI and [
18 F]-FDG-PET imaging, respectively. RNA-Seq, RTK assay, and immunoblotting analysis were used to evaluate gene pathway regulation in vivo and in vitro Analysis of TGII demonstrated significant antitumor effects with cabozantinib compared with regorafenib (average TGII 3.202 vs. 48.48, respectively; P = 0.007). Cabozantinib significantly reduced vascularity and glucose uptake compared with baseline. Gene pathway analysis showed that cabozantinib significantly decreased protein activity involved in glycolysis and upregulated proteins involved in autophagy compared with control, whereas regorafenib did not. The combination of two separate antiautophagy agents, SBI-0206965 and chloroquine, plus cabozantinib increased apoptosis in vitro Cabozantinib demonstrated significant antitumor activity, reduction in tumor vascularity, increased autophagy, and altered cell metabolism compared with regorafenib. Our findings support further evaluation of cabozantinib and combinational approaches targeting autophagy in colorectal cancer. Mol Cancer Ther; 17(10); 2112-22. ©2018 AACR ., (©2018 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
38. Phase Ib Results of the Rational Combination of Selumetinib and Cyclosporin A in Advanced Solid Tumors with an Expansion Cohort in Metastatic Colorectal Cancer.
- Author
-
Krishnamurthy A, Dasari A, Noonan AM, Mehnert JM, Lockhart AC, Leong S, Capasso A, Stein MN, Sanoff HK, Lee JJ, Hansen A, Malhotra U, Rippke S, Gustafson DL, Pitts TM, Ellison K, Davis SL, Messersmith WA, Eckhardt SG, and Lieu CH
- Subjects
- Adolescent, Adult, Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Area Under Curve, Colorectal Neoplasms pathology, Disease-Free Survival, Female, Genes, ras, Humans, Male, Middle Aged, Mutation, Neoplasm Metastasis, Neoplasm Transplantation, Neoplasms drug therapy, Protein Kinase Inhibitors therapeutic use, Signal Transduction, Young Adult, Benzimidazoles administration & dosage, Colorectal Neoplasms drug therapy, Cyclosporine administration & dosage
- Abstract
MEK inhibition is of interest in cancer drug development, but clinical activity in metastatic colorectal cancer (mCRC) has been limited. Preclinical studies demonstrated Wnt pathway overexpression in KRAS -mutant cell lines resistant to the MEK inhibitor, selumetinib. The combination of selumetinib and cyclosporin A, a noncanonical Wnt pathway modulator, demonstrated antitumor activity in mCRC patient-derived xenografts. To translate these results, we conducted a NCI Cancer Therapy Evaluation Program-approved multicenter phase I/IB trial (NCT02188264) of the combination of selumetinib and cyclosporin A. Patients with advanced solid malignancies were treated with the combination of oral selumetinib and cyclosporin A in the dose escalation phase, followed by an expansion cohort of irinotecan and oxaliplatin-refractory mCRC. The expansion cohort utilized a single-agent selumetinib "run-in" to evaluate FZD2 biomarker upregulation and KRAS -WT and KRAS -MT stratification to identify any potential predictors of efficacy. Twenty and 19 patients were enrolled in dose escalation and expansion phases, respectively. The most common adverse events and grade 3/4 toxicities were rash, hypertension, and edema. Three dose-limiting toxicities (grade 3 hypertension, rash, and increased creatinine) were reported. The MTD was selumetinib 75 mg twice daily and cyclosporin A 2 mg/kg twice daily on a 28-day cycle. KRAS stratification did not identify any differences in response between KRAS -WT and KRAS -MT cancers. Two partial responses, 18 stable disease, and 10 progressive disease responses were observed. Combination selumetinib and cyclosporin A is well tolerated, with evidence of activity in mCRC. Future strategies for concept development include identifying better predictors of efficacy and improved Wnt pathway modulation. Significance: These findings translate preclinical studies combining selumetinib and cyclosporin into a phase I first-in-human clinical trial of such a combination in patients with advanced solid malignancies. Cancer Res; 78(18); 5398-407. ©2018 AACR ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
39. Biliverdin Reductase B Dynamics Are Coupled to Coenzyme Binding.
- Author
-
Paukovich N, Xue M, Elder JR, Redzic JS, Blue A, Pike H, Miller BG, Pitts TM, Pollock DD, Hansen K, D'Alessandro A, and Eisenmesser EZ
- Subjects
- Binding Sites, Catalytic Domain, Coenzymes genetics, Coenzymes metabolism, Flavin-Adenine Dinucleotide chemistry, Magnetic Resonance Spectroscopy, Models, Molecular, Mutation, NADP chemistry, NADP metabolism, Oxidoreductases Acting on CH-CH Group Donors metabolism, Protein Conformation, Structure-Activity Relationship, Coenzymes chemistry, Oxidoreductases Acting on CH-CH Group Donors chemistry
- Abstract
Biliverdin reductase B (BLVRB) is a newly identified cellular redox regulator that catalyzes the NADPH-dependent reduction of multiple substrates. Through mass spectrometry analysis, we identified high levels of BLVRB in mature red blood cells, highlighting the importance of BLVRB in redox regulation. The BLVRB conformational changes that occur during conezyme/substrate binding and the role of dynamics in BLVRB function, however, remain unknown. Through a combination of NMR, kinetics, and isothermal titration calorimetry studies, we determined that BLVRB binds its coenzyme 500-fold more tightly than its substrate. While the active site of apo BLVRB is highly dynamic on multiple timescales, active site dynamics are largely quenched within holo BLVRB, in which dynamics are redistributed to other regions of the enzyme. We show that a single point mutation of Arg78➔Ala leads to both an increase in active site micro-millisecond motions and an increase in the microscopic rate constants of coenzyme binding. This demonstrates that altering BLVRB active site dynamics can directly cause a change in functional characteristics. Our studies thus address the solution behavior of apo and holo BLVRB and identify a role of enzyme dynamics in coenzyme binding., (Copyright © 2018. Published by Elsevier Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
40. A phase II clinical trial of the Aurora and angiogenic kinase inhibitor ENMD-2076 for previously treated, advanced, or metastatic triple-negative breast cancer.
- Author
-
Diamond JR, Eckhardt SG, Pitts TM, van Bokhoven A, Aisner D, Gustafson DL, Capasso A, Sams S, Kabos P, Zolman K, Colvin T, Elias AD, Storniolo AM, Schneider BP, Gao D, Tentler JJ, Borges VF, and Miller KD
- Subjects
- Administration, Oral, Adult, Aged, Antineoplastic Agents pharmacokinetics, Aurora Kinase A antagonists & inhibitors, Aurora Kinase A metabolism, Biopsy, Breast pathology, Breast Neoplasms, Male blood, Breast Neoplasms, Male pathology, Diarrhea chemically induced, Diarrhea epidemiology, Disease Progression, Fatigue chemically induced, Fatigue epidemiology, Female, Humans, Hypertension chemically induced, Hypertension epidemiology, Male, Middle Aged, Nausea chemically induced, Nausea epidemiology, Progression-Free Survival, Protein Kinase Inhibitors pharmacokinetics, Pyrazoles pharmacokinetics, Pyrimidines pharmacokinetics, Triple Negative Breast Neoplasms blood, Triple Negative Breast Neoplasms pathology, Antineoplastic Agents therapeutic use, Breast Neoplasms, Male drug therapy, Protein Kinase Inhibitors therapeutic use, Pyrazoles therapeutic use, Pyrimidines therapeutic use, Triple Negative Breast Neoplasms drug therapy
- Abstract
Background: Triple-negative breast cancer (TNBC) remains an aggressive breast cancer subtype with limited treatment options. ENMD-2076 is a small-molecule inhibitor of Aurora and angiogenic kinases with proapoptotic and antiproliferative activity in preclinical models of TNBC., Methods: This dual-institution, single-arm, two-stage, phase II clinical trial enrolled patients with locally advanced or metastatic TNBC previously treated with one to three prior lines of chemotherapy in the advanced setting. Patients were treated with ENMD-2076 250 mg orally once daily with continuous dosing in 4-week cycles until disease progression or unacceptable toxicity occurred. The primary endpoint was 6-month clinical benefit rate (CBR), and secondary endpoints included progression-free survival, pharmacokinetic profile, safety, and biologic correlates in archival and fresh serial tumor biopsies in a subset of patients., Results: Forty-one patients were enrolled. The 6-month CBR was 16.7% (95% CI, 6-32.8%) and included two partial responses. The 4-month CBR was 27.8% (95% CI, 14-45.2%), and the average duration of benefit was 6.5 cycles. Common adverse events included hypertension, fatigue, diarrhea, and nausea. Treatment with ENMD-2076 resulted in a decrease in cellular proliferation and microvessel density and an increase in p53 and p73 expression, consistent with preclinical observations., Conclusions: Single-agent ENMD-2076 treatment resulted in partial response or clinical benefit lasting more than 6 months in 16.7% of patients with pretreated, advanced, or metastatic TNBC. These results support the development of predictive biomarkers using archival and fresh tumor tissue, as well as consideration of mechanism-based combination strategies., Trial Registration: ClinicalTrials.gov, NCT01639248 . Registered on July 12, 2012.
- Published
- 2018
- Full Text
- View/download PDF
41. Development of new preclinical models to advance adrenocortical carcinoma research.
- Author
-
Kiseljak-Vassiliades K, Zhang Y, Bagby SM, Kar A, Pozdeyev N, Xu M, Gowan K, Sharma V, Raeburn CD, Albuja-Cruz M, Jones KL, Fishbein L, Schweppe RE, Somerset H, Pitts TM, Leong S, and Wierman ME
- Subjects
- Adrenal Cortex metabolism, Adrenal Cortex Neoplasms genetics, Adrenal Cortex Neoplasms metabolism, Adrenocortical Carcinoma genetics, Adrenocortical Carcinoma metabolism, Aldosterone metabolism, Cell Line, Tumor, Colorectal Neoplasms, Hereditary Nonpolyposis genetics, Colorectal Neoplasms, Hereditary Nonpolyposis metabolism, Germ-Line Mutation, Humans, Adrenal Cortex pathology, Adrenal Cortex Neoplasms pathology, Adrenocortical Carcinoma pathology, Colorectal Neoplasms, Hereditary Nonpolyposis pathology
- Abstract
Adrenocortical cancer (ACC) is an orphan malignancy that results in heterogeneous clinical phenotypes and molecular genotypes. There are no curative treatments for this deadly cancer with 35% survival at five years. Our understanding of the underlying pathobiology and our ability to test novel therapeutic targets has been limited due to the lack of preclinical models. Here, we report the establishment of two new ACC cell lines and corresponding patient-derived xenograft (PDX) models. CU-ACC1 cell line and PDX were derived from a perinephric metastasis in a patient whose primary tumor secreted aldosterone. CU-ACC2 cell line and PDX were derived from a liver metastasis in a patient with Lynch syndrome. Short tandem repeat profiling confirmed consistent matches between human samples and models. Both exomic and RNA sequencing profiling were performed on the patient samples and the models, and hormonal secretion was evaluated in the new cell lines. RNA sequencing and immunohistochemistry confirmed the expression of adrenal cortex markers in the PDXs and human tumors. The new cell lines replicate two of the known genetic models of ACC. CU-ACC1 cells had a mutation in CTNNB1 and secreted cortisol but not aldosterone. CU-ACC2 cells had a TP53 mutation and loss of MSH2 consistent with the patient's known germline mutation causing Lynch syndrome. Both cell lines can be transfected and transduced with similar growth rates. These new preclinical models of ACC significantly advance the field by allowing investigation of underlying molecular mechanisms of ACC and the ability to test patient-specific therapeutic targets., (© 2018 Society for Endocrinology.)
- Published
- 2018
- Full Text
- View/download PDF
42. Antitumor activity of the polo-like kinase inhibitor, TAK-960, against preclinical models of colorectal cancer.
- Author
-
Klauck PJ, Bagby SM, Capasso A, Bradshaw-Pierce EL, Selby HM, Spreafico A, Tentler JJ, Tan AC, Kim J, Arcaroli JJ, Purkey A, Messersmith WA, Kuida K, Eckhardt SG, and Pitts TM
- Subjects
- 4-Aminobenzoic Acid pharmacology, Animals, Antineoplastic Agents pharmacology, Cell Cycle Proteins metabolism, Cell Line, Tumor, Cell Proliferation drug effects, Colorectal Neoplasms pathology, Female, G2 Phase Cell Cycle Checkpoints drug effects, HCT116 Cells, HT29 Cells, Humans, Mice, Nude, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins metabolism, Tumor Burden drug effects, Polo-Like Kinase 1, Azepines pharmacology, Cell Cycle Proteins antagonists & inhibitors, Colorectal Neoplasms drug therapy, Protein Serine-Threonine Kinases antagonists & inhibitors, Proto-Oncogene Proteins antagonists & inhibitors, Xenograft Model Antitumor Assays
- Abstract
Background: Polo-like kinase 1 (Plk1) is a serine/threonine kinase that is a key regulator of multiple stages of mitotic progression. Plk1 is upregulated in many tumor types including colorectal cancer (CRC) and portends a poor prognosis. TAK-960 is an ATP-competitive Plk1 inhibitor that has demonstrated efficacy across a broad range of cancer cell lines, including CRC. In this study, we investigated the activity of TAK-960 against a large collection of CRC models including 55 cell lines and 18 patient-derived xenografts., Methods: Fifty-five CRC cell lines and 18 PDX models were exposed to TAK-960 and evaluated for proliferation (IC
50 ) and Tumor Growth Inhibition Index, respectively. Additionally, 2 KRAS wild type and 2 KRAS mutant PDX models were treated with TAK-960 as single agent or in combination with cetuximab or irinotecan. TAK-960 mechanism of action was elucidated through immunoblotting and cell cycle analysis., Results: CRC cell lines demonstrated a variable anti-proliferative response to TAK-960 with IC50 values ranging from 0.001 to > 0.75 μmol/L. Anti-proliferative effects were sustained after removal of drug. Following TAK-960 treatment a highly variable accumulation of mitotic (indicating cell cycle arrest) and apoptotic markers was observed. Cell cycle analysis demonstrated that TAK-960 treatment induced G2/M arrest and polyploidy. Six out of the eighteen PDX models responded to single agent TAK-960 therapy (TGII< 20). The addition of TAK-960 to standard of care chemotherapy resulted in largely additive antitumor effects., Conclusion: TAK-960 is an active anti-proliferative agent against CRC cell lines and PDX models. Collectively, these data suggest that TAK-960 may be of therapeutic benefit alone or in combination with other agents, although future work should focus on the development of predictive biomarkers and hypothesis-driven rational combinations.- Published
- 2018
- Full Text
- View/download PDF
43. Evaluation of TAK-264, an Antibody-Drug Conjugate in Pancreatic Cancer Cell Lines and Patient-Derived Xenograft Models.
- Author
-
Schreiber AR, Nguyen A, Bagby SM, Arcaroli JJ, Yacob BW, Quackenbush K, Guy JL, Crowell T, Stringer B, Danaee H, Kalebic T, Messersmith WA, and Pitts TM
- Abstract
Background: Antibody-drug conjugates (ADCs) are an emerging technology consisting of an antibody, linker, and toxic agent, which have the potential to offer a targeted therapeutic approach. A novel target recently explored for the treatment of pancreatic cancer is guanylyl cyclase C (GCC). The objective of this study was to determine the anti-tumorigenic activity of TAK-264, an investigational ADC consisting of an antibody targeting GCC linked to a monomethyl auristatin E payload via a peptide linker., Methods: The antiproliferative effects of TAK-264 assessed in a panel of eleven pancreatic cancer cell lines. Additionally, ten unique pancreatic ductal adenocarcinoma cancer patient-derived xenograft models were treated with TAK-264 and the efficacy was determined. Baseline levels of GCC were analyzed on PDX models and cell lines. Immunoblotting was performed to evaluate the effects of TAK-264 on downstream effectors., Results: GCC protein expression was analyzed by immunoblotting in both normal and tumor tissue; marked increase in GCC expression was observed in tumor tissue. The in vitro experiments demonstrated a range of responses to TAK-264. Eight of the ten PDAC PDX models treated with TAK-264 demonstrated a statistically significant tumor growth inhibition. Immunoblotting demonstrated an increase in phosphorylated-HistoneH3 in both responsive and less responsive cell lines and PDAC PDX models treated with TAK-264. There was no correlation between baseline levels of GCC and response in either PDX or cell line models., Conclusion: TAK-264 has shown suppression activity in pancreatic cancer cell lines and in pancreatic PDX models. These findings support further investigation of ADC targeting GCC., Competing Interests: CONFLICT OF INTEREST Potential conflict of interest are authors TK, BS, HD, TC who are employees of Takeda Limited. Takeda Pharmaceuticals partially funded this project and had no role in study design and decision to publish. The funders had no role in data collection and analysis or preparation of the manuscript. Authors ARS, AN, SMB, JJA, BY, KQ, JLG, WAM, TMP have no conflicts of interest.
- Published
- 2018
- Full Text
- View/download PDF
44. ALK Inhibitor Response in Melanomas Expressing EML4-ALK Fusions and Alternate ALK Isoforms.
- Author
-
Couts KL, Bemis J, Turner JA, Bagby SM, Murphy D, Christiansen J, Hintzsche JD, Le A, Pitts TM, Wells K, Applegate A, Amato C, Multani P, Chow-Maneval E, Tentler JJ, Shellman YG, Rioth MJ, Tan AC, Gonzalez R, Medina T, Doebele RC, and Robinson WA
- Subjects
- Animals, Female, Humans, Melanoma pathology, Mice, Mice, Nude, Middle Aged, Protein Isoforms, Xenograft Model Antitumor Assays, Melanoma genetics, Oncogene Proteins, Fusion genetics, Oncogene Proteins, Fusion metabolism
- Abstract
Oncogenic ALK fusions occur in several types of cancer and can be effectively treated with ALK inhibitors; however, ALK fusions and treatment response have not been characterized in malignant melanomas. Recently, a novel isoform of ALK ( ALK
ATI ) was reported in 11% of melanomas but the response of melanomas expressing ALKATI to ALK inhibition has not been well characterized. We analyzed 45 melanoma patient-derived xenograft models for ALK mRNA and protein expression. ALK expression was identified in 11 of 45 (24.4%) melanomas. Ten melanomas express wild-type (wt) ALK and/or ALKATI and one mucosal melanoma expresses multiple novel EML4-ALK fusion variants. Melanoma cells expressing different ALK variants were tested for response to ALK inhibitors. Whereas the melanoma expressing EML4-ALK were sensitive to ALK inhibitors in vitro and in vivo , the melanomas expressing wt ALK or ALKATI were not sensitive to ALK inhibitors. In addition, a patient with mucosal melanoma expressing ALKATI was treated with an ALK/ROS1/TRK inhibitor (entrectinib) on a phase I trial but did not respond. Our results demonstrate ALK fusions occur in malignant melanomas and respond to targeted therapy, whereas melanomas expressing ALKATI do not respond to ALK inhibitors. Targeting ALK fusions is an effective therapeutic option for a subset of melanoma patients, but additional clinical studies are needed to determine the efficacy of targeted therapies in melanomas expressing wt ALK or ALKATI Mol Cancer Ther; 17(1); 222-31. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
45. The novel ATM inhibitor (AZ31) enhances antitumor activity in patient derived xenografts that are resistant to irinotecan monotherapy.
- Author
-
Greene J, Nguyen A, Bagby SM, Jones GN, Tai WM, Quackenbush KS, Schreiber A, Messersmith WA, Devaraj KM, Blatchford P, Eckhardt SG, Cadogan EB, Hughes GD, Smith A, Pitts TM, and Arcaroli JJ
- Abstract
Irinotecan, a standard of care therapy for CRC, elicits cytotoxic effects by generating double strand breaks resulting in DNA damage. The activation of the ATM pathway plays a fundamental role in regulating the cellular response and repair to DNA damage. The objective of this preclinical study was to determine whether ATM inhibition would enhance sensitivity to irinotecan treatment. Treatment effects of AZ31, irinotecan or AZ31 + irinotecan were investigated in CRC cell lines and CRC patient derived xenografts. Activation of ATM and downstream targets p-RAD50 and p-H2AX were evaluated by immunohistochemistry. Combinational effects were demonstrated in 4 out of 8 CRC explants. Interestingly, each of the combinational sensitive CRC PDX models were shown to be more resistant to irinotecan single agent therapy. Treatment with irinotecan significantly elevated the ATM pathway evident by an increase in the activation of H2AX and RAD50. Combinational therapy reduced the activation of H2AX and RAD50 when compared to irinotecan alone in the combination sensitive CRC098. AZ31 + irinotecan was effective at reducing tumor growth in tumors that exhibited resistance to irinotecan in our CRC PDX model. These findings support further investigation of this combinational therapy for the treatment of CRC patients., Competing Interests: CONFLICTS OF INTEREST None.
- Published
- 2017
- Full Text
- View/download PDF
46. Evaluation of the efficacy of dasatinib, a Src/Abl inhibitor, in colorectal cancer cell lines and explant mouse model.
- Author
-
Scott AJ, Song EK, Bagby S, Purkey A, McCarter M, Gajdos C, Quackenbush KS, Cross B, Pitts TM, Tan AC, Eckhardt SG, Fenton H, Arcaroli J, and Messersmith WA
- Subjects
- Animals, Apoptosis drug effects, Cell Cycle drug effects, Cell Line, Tumor, Humans, Mice, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Dasatinib pharmacology, Oncogene Proteins v-abl antagonists & inhibitors, Protein Kinase Inhibitors pharmacology, src-Family Kinases antagonists & inhibitors
- Abstract
Background: Dysregulation of the Src pathway has been shown to be important at various stages of cancer. Dasatinib is a potent Src/Abl inhibitor and has demonstrated to have anti-proliferative and anti-invasive activity in many preclinical models. The objective of this study was to determine the anti-tumor activity of dasatinib using in vitro and in vivo preclinical colorectal (CRC) models., Methods: CRC cell lines and patient-derived tumor explant (PDX) models were used to investigate the efficacy of dasatinib. We treated 50 CRC cell lines with dasatinib for 72 hours and proliferation was assayed by a sulforhodamine B (SRB) assay; an IC50 ≤ 0.08 μmol/L was considered sensitive. We treated 17 patient-derived CRC explants with dasatinib (50 mg/kg/day, administered once-daily) for 28 days to determine in vivo efficacy. Tumor growth inhibition (TGI) ≥ 50% was considered sensitive., Results: We found that 8 out of 50 CRC cell lines reached an IC50 ≤ 0.08 μmol/L with dasatinib treatment. In addition, of 17 CRC explants grown in the xenograft mouse model, 2 showed sensitivity to dasatinib. The anti-tumor effects observed in this study were a result of G1 cell cycle arrest as the dasatinib sensitive CRC cell lines exhibited G1 inhibition. Moreover, those CRC cell lines that were responsive (0.08 μmol/L) to treatment demonstrated a significant baseline increase in Src and FAK gene expression., Conclusion: Dasatinib demonstrated significant anti-proliferative activity in a subset of CRC cell lines in vitro, especially in those with increased Src expression at baseline, but only showed modest efficacy in CRC explants. Dasatinib is currently being studied in combination with chemotherapy in patients with advanced CRC, as its use as a single agent appears limited.
- Published
- 2017
- Full Text
- View/download PDF
47. Efficacy and Molecular Mechanisms of Differentiated Response to the Aurora and Angiogenic Kinase Inhibitor ENMD-2076 in Preclinical Models of p53-Mutated Triple-Negative Breast Cancer.
- Author
-
Ionkina AA, Tentler JJ, Kim J, Capasso A, Pitts TM, Ryall KA, Howison RR, Kabos P, Sartorius CA, Tan AC, Eckhardt SG, and Diamond JR
- Abstract
Purpose: Triple-negative breast cancer (TNBC) is a subtype associated with poor prognosis and for which there are limited therapeutic options. The purpose of this study was to evaluate the efficacy of ENMD-2076 in p53-mutated TNBC patient-derived xenograft (PDX) models and describe patterns of terminal cell fate in models demonstrating sensitivity, intrinsic resistance, and acquired resistance to ENMD-2076., Experimental Design: p53-mutated, TNBC PDX models were treated with ENMD-2076 and evaluated for mechanisms of sensitivity or resistance to treatment. Correlative tissue testing was performed on tumor tissue to assess for markers of proliferation, apoptosis, senescence, and pathways of resistance after treatment and at the time of acquired resistance., Results: Sensitivity to ENMD-2076 200 mg/kg daily was associated with induction of apoptosis while models exhibiting intrinsic or acquired resistance to treatment presented with a senescent phenotype. Response to ENMD-2076 was accompanied by an increase in p53 and p73 levels, even within the background of mutant p53. Treatment with ENMD-2076 resulted in a decrease in pAurA and an increase in pHH3. We observed a TNBC subtype switch from the luminal androgen receptor to the basal-like subtype at acquired resistance., Conclusion: ENMD-2076 has antitumor activity in preclinical models of p53-mutated TNBC. Increased levels of p53 and p73 correlated with sensitivity whereas senescence was associated with resistance to ENMD-2076. The novel finding of a TNBC subtype switch at time of acquired resistance may provide mechanistic insights into the biologic effects of selective pressure of anticancer treatments on TNBC. ENMD-2076 is currently being evaluated in a Phase 2 clinical trial in patients with metastatic, previously treated TNBC where these biologic correlates can be further explored.
- Published
- 2017
- Full Text
- View/download PDF
48. Procedure for Horizontal Transfer of Patient-Derived Xenograft Tumors to Eliminate Corynebacterium bovis .
- Author
-
Manuel CA, Bagby SM, Reisinger JA, Pugazhenthi U, Pitts TM, Keysar SB, Arcaroli JJ, and Leszczynski JK
- Subjects
- Animals, Corynebacterium Infections microbiology, Humans, Mice, Mice, Nude, Neoplasms, Experimental pathology, Corynebacterium classification, Corynebacterium Infections prevention & control, Heterografts microbiology, Neoplasms, Experimental microbiology
- Abstract
Human patient-derived xenograft (PDX) tumors, propagated in immunodeficient mice, are rapidly growing in use as a model for cancer research. Horizontal transfer between mice, without in vitro cell culture, allows these tumors to retain many of their unique characteristics from their individual patient of origin. However, the immunodeficient mouse strains used to grow these tumors are susceptible to numerous opportunistic pathogens, including Corynebacterium bovis. At our institution, 2 in vivo tumor banks of PDX tumors had been maintained within nude mouse colonies enzootically infected with C. bovis. Elimination of C. bovis from these colonies required the aseptic harvest and horizontal transfer of tumor tissue between infected and naïve recipient mice without cross-contamination. Out of necessity, we developed a standard operating procedure using enhancements to traditional aseptic surgical technique with concurrent application of both procedural and physical barriers to prevent C. bovis transmission. By using these methods, all 61 unique PDX tumor models were successfully harvested from C. bovis-infected mice and transferred into recipient mice without transmission of infection. Our data demonstrate that, in situations where C. bovis-free colonies can be established and maintained, this procedure can successfully be used to eliminate C. bovis from an in vivo tumor bank of valuable PDX tumors.
- Published
- 2017
49. Targeting the protein ubiquitination machinery in melanoma by the NEDD8-activating enzyme inhibitor pevonedistat (MLN4924).
- Author
-
Wong KM, Micel LN, Selby HM, Tan AC, Pitts TM, Bagby SM, Spreafico A, Klauck PJ, Blakemore SJ, Smith PF, McDonald A, Berger A, Tentler JJ, and Eckhardt SG
- Subjects
- Apoptosis drug effects, Cell Line, Tumor, Cell Survival drug effects, Gene Expression Regulation, Neoplastic drug effects, Humans, Melanoma genetics, Melanoma metabolism, Ubiquitin-Activating Enzymes metabolism, Ubiquitination drug effects, Antineoplastic Agents pharmacology, Cyclopentanes pharmacology, Melanoma drug therapy, Pyrimidines pharmacology, Ubiquitin-Activating Enzymes antagonists & inhibitors
- Abstract
Background The neddylation pathway conjugates NEDD8 to cullin-RING ligases and controls the proteasomal degradation of specific proteins involved in essential cell processes. Pevonedistat (MLN4924) is a selective small molecule targeting the NEDD8-activating enzyme (NAE) and inhibits an early step in neddylation, resulting in DNA re-replication, cell cycle arrest and death. We investigated the anti-tumor potential of pevonedistat in preclinical models of melanoma. Methods Melanoma cell lines and patient-derived tumor xenografts (PDTX) treated with pevonedistat were assessed for viability/apoptosis and tumor growth, respectively, to identify sensitive/resistant models. Gene expression microarray and gene set enrichment analyses were performed in cell lines to determine the expression profiles and pathways of sensitivity/resistance. Pharmacodynamic changes in treated-PDTX were also characterized. Results Pevonedistat effectively inhibited cell viability (IC
50 < 0.3 μM) and induced apoptosis in a subset of melanoma cell lines. Sensitive and resistant cell lines exhibited distinct gene expression profiles; sensitive models were enriched for genes involved in DNA repair, replication and cell cycle regulation, while immune response and cell adhesion pathways were upregulated in resistant models. Pevonedistat also reduced tumor growth in melanoma cell line xenografts and PDTX with variable responses. An accumulation of pevonedistat-NEDD8 adduct and CDT1 was observed in sensitive tumors consistent with its mechanism of action. Conclusions This study provided preclinical evidence that NAE inhibition by pevonedistat has anti-tumor activity in melanoma and supports the clinical benefits observed in recent Phase 1 trials of this drug in melanoma patients. Further investigations are warranted to develop rational combinations and determine predictive biomarkers of pevonedistat.- Published
- 2017
- Full Text
- View/download PDF
50. Development and Maintenance of a Preclinical Patient Derived Tumor Xenograft Model for the Investigation of Novel Anti-Cancer Therapies.
- Author
-
Bagby S, Messersmith WA, Pitts TM, Capasso A, Varella-Garcia M, Klauck PJ, Kim J, Tan AC, Eckhardt SG, Tentler JJ, and Arcaroli J
- Subjects
- Animals, Antineoplastic Agents therapeutic use, Heterografts, Humans, Mice, Mice, Nude, Neoplasm Metastasis, Neoplasms, Transplantation, Heterologous, Drug Discovery, Xenograft Model Antitumor Assays
- Abstract
Patient derived tumor xenograft (PDTX) models provide a necessary platform in facilitating anti-cancer drug development prior to human trials. Human tumor pieces are injected subcutaneously into athymic nude mice (immunocompromised, T cell deficient) to create a bank of tumors and subsequently are passaged into different generations of mice in order to maintain these tumors from patients. Importantly, cellular heterogeneity of the original tumor is closely emulated in this model, which provides a more clinically relevant model for evaluation of drug efficacy studies (single agent and combination), biomarker analysis, resistant pathways and cancer stem cell biology. Some limitations of the PDTX model include the replacement of the human stroma with mouse stroma after the first generation in mice, inability to investigate treatment effects on metastasis due to the subcutaneous injections of the tumors, and the lack of evaluation of immunotherapies due to the use of immunocompromised mice. However, even with these limitations, the PDTX model provides a powerful preclinical platform in the drug discovery process.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.