Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where David Monsma is active.

Publication


Featured researches published by David Monsma.


Molecular Cancer Research | 2005

Analysis of Tumor-Host Interactions by Gene Expression Profiling of Lung Adenocarcinoma Xenografts Identifies Genes Involved in Tumor Formation

Chad J. Creighton; Jennifer L. Bromberg-White; David E. Misek; David Monsma; Frank Brichory; Rork Kuick; Thomas J. Giordano; Weimin Gao; Gilbert S. Omenn; Craig P. Webb; Samir M. Hanash

Tumor cell lines are relied on extensively for cancer investigations, yet cultured cells in an in vitro environment differ considerably in behavior compared with those of the same cancer cells that proliferate and form tumors in vivo. To uncover gene expression changes related to tumor formation, gene expression profiles of human lung adenocarcinoma (A549) cells grown as lung tumors in immune-compromised mice were compared with profiles of the same cells grown in vitro. Additionally, profiles of uninvolved adjacent mouse tissue were determined. A profound interplay between cancer cells and the host was shown that affected a complex protein interaction network involving processes of extracellular interaction, growth factor signaling, hemostasis, immune response, and transcriptional regulation. Growth in vivo of A549 cells, which carry an activating k-ras mutation, induced changes in gene expression that corresponded highly to a pattern characteristic of human lung tumors with k-ras mutation. Cytokines interleukin-4, interleukin-6, and IFN-γ each induced distinct in vitro genomic responses in cancer cells that emulated many of the changes in gene expression observed in vivo. Genes that were both selectively induced in vivo and overexpressed in human lung adenocarcinoma tumors included CSPG2, which has not been associated previously with tumor formation. Knockdown in A549 of CSPG2 by RNA interference significantly inhibited tumor growth in vivo but not in vitro. Thus, analysis of tumor xenografts by gene expression profiling has the potential for identifying genes involved in tumor development that may not be expressed in cancer cells grown in vitro.


Journal of Translational Medicine | 2012

Genomic characterization of explant tumorgraft models derived from fresh patient tumor tissue

David Monsma; Noel R. Monks; David Cherba; Dawna Dylewski; Emily Eugster; Hailey Jahn; Sujata Srikanth; Stephanie B. Scott; Patrick J. Richardson; Robin E. Everts; Aleksandr Ishkin; Yuri Nikolsky; James H. Resau; Robert E. Sigler; Brian J. Nickoloff; Craig P. Webb

BackgroundThere is resurgence within drug and biomarker development communities for the use of primary tumorgraft models as improved predictors of patient tumor response to novel therapeutic strategies. Despite perceived advantages over cell line derived xenograft models, there is limited data comparing the genotype and phenotype of tumorgrafts to the donor patient tumor, limiting the determination of molecular relevance of the tumorgraft model. This report directly compares the genomic characteristics of patient tumors and the derived tumorgraft models, including gene expression, and oncogenic mutation status.MethodsFresh tumor tissues from 182 cancer patients were implanted subcutaneously into immune-compromised mice for the development of primary patient tumorgraft models. Histological assessment was performed on both patient tumors and the resulting tumorgraft models. Somatic mutations in key oncogenes and gene expression levels of resulting tumorgrafts were compared to the matched patient tumors using the OncoCarta (Sequenom, San Diego, CA) and human gene microarray (Affymetrix, Santa Clara, CA) platforms respectively. The genomic stability of the established tumorgrafts was assessed across serial in vivo generations in a representative subset of models. The genomes of patient tumors that formed tumorgrafts were compared to those that did not to identify the possible molecular basis to successful engraftment or rejection.ResultsFresh tumor tissues from 182 cancer patients were implanted into immune-compromised mice with forty-nine tumorgraft models that have been successfully established, exhibiting strong histological and genomic fidelity to the originating patient tumors. Comparison of the transcriptomes and oncogenic mutations between the tumorgrafts and the matched patient tumors were found to be stable across four tumorgraft generations. Not only did the various tumors retain the differentiation pattern, but supporting stromal elements were preserved. Those genes down-regulated specifically in tumorgrafts were enriched in biological pathways involved in host immune response, consistent with the immune deficiency status of the host. Patient tumors that successfully formed tumorgrafts were enriched for cell signaling, cell cycle, and cytoskeleton pathways and exhibited evidence of reduced immunogenicity.ConclusionsThe preservation of the patient’s tumor genomic profile and tumor microenvironment supports the view that primary patient tumorgrafts provide a relevant model to support the translation of new therapeutic strategies and personalized medicine approaches in oncology.


DNA and Cell Biology | 2011

In vivo Safety and Antitumor Efficacy of Bifunctional Small Hairpin RNAs Specific for the Human Stathmin 1 Oncoprotein

Anagha P. Phadke; Chris M. Jay; Zhaohui Wang; Salina Chen; Shengnan Liu; Courtney Haddock; Padmasini Kumar; Beena O. Pappen; Donald Rao; Nancy Smyth Templeton; Egeenee Q. Daniels; Craig P. Webb; David Monsma; Stephanie B. Scott; Dawna Dylewski; Hermann B. Frieboes; F.C. Brunicardi; Neil Senzer; Phillip B. Maples; John Nemunaitis; Alex W. Tong

Bifunctional small hairpin RNAs (bi-shRNAs) are functional miRNA/siRNA composites that are optimized for posttranscriptional gene silencing through concurrent mRNA cleavage-dependent and -independent mechanisms (Rao et al., 2010 ). We have generated a novel bi-shRNA using the miR30 scaffold that is highly effective for knockdown of human stathmin (STMN1) mRNA. STMN1 overexpression well documented in human solid cancers correlates with their poor prognosis. Transfection with the bi-shSTMN1-encoding expression plasmid (pbi-shSTMN1) markedly reduced CCL-247 human colorectal cancer and SK-Mel-28 melanoma cell growth in vitro (Rao et al., 2010 ). We now examine in vivo the antitumor efficacy of this RNA interference-based approach with human tumor xenografted athymic mice. A single intratumoral (IT) injection of pbi-shSTMN1 (8 μg) reduced CCL-247 tumor xenograft growth by 44% at 7 days when delivered as a 1,2-dioleoyl-3-trimethyl-ammoniopropane:cholesterol liposomal complex. Extended growth reductions (57% at day 15; p < 0.05) were achieved with three daily treatments of the same construct. STMN1 protein reduction was confirmed by immunoblot analysis. IT treatments with pbi-shSTMN1 similarly inhibited the growth of tumorgrafts derived from low-passage primary melanoma (≥70% reduction for 2 weeks) and abrogated osteosarcoma tumorgraft growth, with the mature bi-shRNA effector molecule detectable for up to 16 days after last injection. Antitumor efficacy was evident for up to 25 days posttreatment in the melanoma tumorgraft model. The maximum tolerated dose by IT injection of >92 μg (Human equivalent dose [HED] of >0.3 mg/kg) in CCL-247 tumor xenograft-bearing athymic mice was ∼10-fold higher than the extrapolated IC(50) of 9 μg (HED of 0.03 mg/kg). Healthy, immunocompetent rats were used as biorelevant models for systemic safety assessments. The observed maximum tolerated dose of <100 μg for intravenously injected pbi-shSTMN1 (mouse equivalent of <26.5 μg; HED of <0.09 mg/kg) confirmed systemic safety of the therapeutic dose, hence supporting early-phase assessments of clinical safety and preliminary efficacy.


Pediatric Blood & Cancer | 2014

Using a rhabdomyosarcoma patient‐derived xenograft to examine precision medicine approaches and model acquired resistance

David Monsma; David Cherba; Patrick J. Richardson; Sean Vance; Sanjeet Rangarajan; Dawna Dylewski; Emily Eugster; Stephanie B. Scott; Nicole L. Beuschel; Paula J. Davidson; Richard Axtell; Deanna Mitchell; Eric Lester; Joseph J. Junewick; Craig P. Webb; Noel R. Monks

Precision (Personalized) medicine has the potential to revolutionize patient health care especially for many cancers where the fundamental disease etiology remains either elusive or has no available therapy. Here we outline a study in alveolar rhabdomyosarcoma, in which we use gene expression profiling and a series of drug prediction algorithms combined with a matched patient‐derived xenograft (PDX) model to test bioinformatically predicted therapies.


Cancer Chemotherapy and Pharmacology | 2014

Development and antitumor activity of a BCL-2 targeted single-stranded DNA oligonucleotide

Wendi V. Rodrigueza; Michael J. Woolliscroft; Abdul Shukkur Ebrahim; Robert Forgey; Patrick McGovren; Gerold Endert; Andreas Wagner; Danielle Holewa; Amro Aboukameel; Richard D. Gill; Charles L. Bisgaier; Richard A. Messmann; Christopher E. Whitehead; Elzbieta Izbicka; Robert T. Streeper; Michael Wick; Gabriela Stiegler; C. A. Stein; David Monsma; Craig P. Webb; Mina Patel Sooch; Steffen Panzner; Ramzi M. Mohammad; Neal Clifford Goodwin; Ayad Al-Katib

PNT100 is a 24-base, chemically unmodified DNA oligonucleotide sequence that is complementary to a region upstream of the BCL-2 gene. Exposure of tumor cells to PNT100 results in suppression of proliferation and cell death by a process called DNA interference. PNT2258 is PNT100 that is encapsulated in protective amphoteric liposomes developed to efficiently encapsulate the PNT100 oligonucleotide, provide enhanced serum stability, optimized pharmacokinetic properties and antitumor activity of the nanoparticle both in vivo and in vitro. PNT2258 demonstrates broad antitumor activity against BCL-2-driven WSU-DLCL2 lymphoma, highly resistant A375 melanoma, PC-3 prostate, and Daudi-Burkitt’s lymphoma xenografts. The sequence specificity of PNT100 was demonstrated against three control sequences (scrambled, mismatched, and reverse complement) all encapsulated in a lipid formulation with identical particle characteristics, and control sequences did not demonstrate antiproliferative activity in vivo or in vitro. PNT2258 is currently undergoing clinical testing to evaluate safety and antitumor activity in patients with recurrent or refractory non-Hodgkin’s lymphoma and additional studies are planned.


Journal of Translational Medicine | 2015

Anoikis-resistant subpopulations of human osteosarcoma display significant chemoresistance and are sensitive to targeted epigenetic therapies predicted by expression profiling

Jessica M Foley; Donald J. Scholten; Noel R. Monks; David Cherba; David Monsma; Paula Davidson; Dawna Dylewski; Karl Dykema; Mary E. Winn; Matthew R. Steensma

BackgroundOsteosarcoma (OS) is the most common type of solid bone cancer, with latent metastasis being a typical mode of disease progression and a major contributor to poor prognosis. For this to occur, cells must resist anoikis and be able to recapitulate tumorigenesis in a foreign microenvironment. Finding novel approaches to treat osteosarcoma and target those cell subpopulations that possess the ability to resist anoikis and contribute to metastatic disease is imperative. Here we investigate anchorage-independent (AI) cell growth as a model to better characterize anoikis resistance in human osteosarcoma while using an expression profiling approach to identify and test targetable signaling pathways.MethodsEstablished human OS cell lines and patient-derived human OS cell isolates were subjected to growth in either adherent or AI conditions using Ultra-Low Attachment plates in identical media conditions. Growth rate was assessed using cell doubling times and chemoresistance was assessed by determining cell viability in response to a serial dilution of either doxorubicin or cisplatin. Gene expression differences were examined using quantitative reverse-transcription PCR and microarray with principal component and pathway analysis. In-vivo OS xenografts were generated by either subcutaneous or intratibial injection of adherent or AI human OS cells into athymic nude mice. Statistical significance was determined using student’s t-tests with significance set at α = 0.05.ResultsWe show that AI growth results in a global gene expression profile change accompanied by significant chemoresistance (up to 75 fold, p < 0.05). AI cells demonstrate alteration of key mediators of mesenchymal differentiation (β-catenin, Runx2), stemness (Sox2), proliferation (c-myc, Akt), and epigenetic regulation (HDAC class 1). AI cells were equally tumorigenic as their adherent counterparts, but showed a significantly decreased rate of growth in-vitro and in-vivo (p < 0.05). Treatment with the pan-histone deacetylase inhibitor vorinostat and the DNA methyltransferase inhibitor 5-azacytidine mitigated AI growth, while 5-azacytidine sensitized anoikis-resistant cells to doxorubicin (p < 0.05).ConclusionsThese data demonstrate remarkable plasticity in anoikis-resistant human osteosarcoma subpopulations accompanied by a rapid development of chemoresistance and altered growth rates mirroring the early stages of latent metastasis. Targeting epigenetic regulation of this process may be a viable therapeutic strategy.


Archive | 2018

Patient-Derived Xenograft Models of Colorectal Cancer: Procedures for Engraftment and Propagation

Danielle M. Burgenske; David Monsma; Jeffrey P. MacKeigan

Preclinical compounds tested in animal models often demonstrate limited efficacy when transitioned into patients. As a result, individuals are assigned to treatment regimens that may be ineffective at treating their disease. The development of more clinically relevant models, such as patient-derived xenografts (PDXs), will (1) more completely mimic the human condition and (2) more accurately predict tumor responses to previously untested therapeutics.PDX models are clinically relevant as tumor tissue is implanted directly from human donor to the mouse recipient. Therefore, these models prevent cell population selection, intentional or unintentional, as the human tissue adapts to an in vitro, two-dimensional environment prior to implantation into a three-dimensional in vivo murine host. Often, cell heterogeneity and tumor architecture can be maintained from human to the PDX model in the mouse. This protocol describes the engraftment and propagation processes for establishing colorectal (CRC) PDX models in mice, using tumor tissue from human subjects.


Molecular Cancer Therapeutics | 2015

Abstract B50: Targeting KRASG12D colorectal cancer with combined inhibition of PI3K/mTOR and MAPK signaling

Danielle M. Burgenske; David Monsma; Dawna Dylewski; Stephanie B. Scott; Aaron Sayfie; Donald Kim; Martin Luchtefeld; Katie R. Martin; Paul Stephenson; Galen Hostetter; Nadav Dujovny; Jeffrey P. MacKeigan

Compounds validated in preclinical models frequently exhibit limited efficacy when transitioned into human trials. As a result, many patients are stratified into treatment regimens that have little impact on their disease. With the goal of establishing preclinical models that can more accurately predict tumor responses, there has been a recent emphasis to develop patient-derived xenograft (PDX) models. We strove to develop new PDX models of colorectal cancer (CRC) to test the feasibility of targeted therapy using small molecule inhibitors. Following transplantation of patient tumor specimens into athymic nude mice, sixteen PDX models were successfully established. Common somatic mutations were determined using custom gene panels and targeted with appropriate inhibitors against PI3K/AKT, mTOR and/or MAPK signaling. In four independent models, single agent therapies against PI3K and mTOR had little impact on tumor growth. Conversely, robust declines in tumor burden were universally observed in all models with MEK inhibition with the exception of one, a KRAS G12D mutant model. Within this model, tumor response was achieved only with dual inhibition of mTOR and MAPK signaling. Given the unmet clinical need to treat tumors with aberrant KRAS signaling, these results encourage further investigation into combination treatment strategies with these small molecule inhibitors. Citation Format: Danielle Burgenske, David Monsma, Dawna Dylewski, Stephanie Scott, Aaron Sayfie, Donald Kim, Martin Luchtefeld, Katie Martin, Paul Stephenson, Galen Hostetter, Nadav Dujovny, Jeffrey MacKeigan. Targeting KRASG12D colorectal cancer with combined inhibition of PI3K/mTOR and MAPK signaling. [abstract]. In: Proceedings of the AACR Special Conference: Targeting the PI3K-mTOR Network in Cancer; Sep 14-17, 2014; Philadelphia, PA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(7 Suppl):Abstract nr B50.


Cancer Epidemiology, Biomarkers & Prevention | 2015

Abstract PR09: Creating a comprehensive patient-derived xenograft (PDX) bank to represent racial disparities in triple-negative breast cancer (TNBC)

Tahra Luther; Evelyn Jiagge; Michael T. Lewis; David Monsma; Craig P. Webb; Suling Liu; Hasan Korkaya; Sean P. McDermott; Shawn G. Clouthier; Lisa A. Newman; Dafydd G. Thomas; Max S. Wicha

Background: Triple negative breast cancer (TNBC) is negative for estrogen receptor (ER), progesterone receptor (PR), and HER2 (human epidermal growth factor receptor 2) and known to be highly aggressive. TNBC carries poor prognosis in all ethnicities but it is especially prevalent in women of African descent, next most prevalent in African American women, and then to a lesser degree in Caucasian women. Cancer cell lines from various ethnicities have been used to study the nature of these tumors, however, PDX models more accurately reflect the characteristics of the original human tumors. To study this aggressive form of breast cancer amongst ethnicities, including up-regulated and down-regulated genes that play major roles across these tumors, we created a multi-ethnic cohort of TNBC PDXs which allows us to model TNBC disease disparities among these populations. Methods: Several trips to Ghana, Africa (Komfo Anoyke Teaching Hospital (KATH)) were made to procure tumors from African women presenting with breast cancer. We created a protocol to successfully freeze and transport tumors back to the University of Michigan, with very limited research supplies available at KATH. Once back at the University of Michigan, tumor pieces were quickly thawed, minced into small fragments and implanted orthotopically into the fat pads of NOD/SCID IL2R gamma null (NSG) mice. For African American and Caucasian PDX development, we collected tumor specimens from lumpectomies performed at the University of Michigan hospital, Baylor College of Medicine, and Van Andel Research Institute and implanted freshly minced tumor pieces into the fat pads of NSG mice. Once the tumors reached at least one centimeter in size, mice were humanely euthanized and their tumors and organs were removed. Samples of each PDX and all organs were formalin-fixed and paraffin-embedded so expression of ER, PR and HER2 receptor status could be confirmed and cancer stem cell populations assessed, and organ metastasis analyzed, by immunohistochemistry. Results: To date we have successfully established (defined by three or more passages) six African, five African American, and five Caucasian TNBC PDXs. We collected TNBC tumors from fifty-six women, of which forty-two tumors were implanted into mice and twenty four PDXs were generated (this total also includes other ethnicities in addition to the three we are looking at in this particular study). Furthermore, we created an inclusive database which indicates those PDXs that grow as tumorspheres to not only study the characteristics and cancer stem cell properties of each of these tumors and compare them based on ethnicity, but also to determine which models are best for investigational drug studies. Conclusions: We developed a method of orthotopic implantation amenable to PDX generation, even for tumor samples collected abroad, which allow us to accurately model TNBC in different ethnicities. These TNBC PDXs afford a more thorough examination of cancer stem cell properties driving aggressiveness of TNBC among different ethnicities and can inform potential drug therapies to treat TNBC and eliminate metastases. This abstract was also presented as Poster A69. Citation Format: Tahra Kaur Luther, Evelyn Jiagge, Michael T. Lewis, David Monsma, Craig Webb, Suling Liu, Hasan Korkaya, Sean McDermott, Shawn G. Clouthier, Lisa Newman, Dafydd Thomas, Max S. Wicha. Creating a comprehensive patient-derived xenograft (PDX) bank to represent racial disparities in triple-negative breast cancer (TNBC). [abstract]. In: Proceedings of the Seventh AACR Conference on The Science of Health Disparities in Racial/Ethnic Minorities and the Medically Underserved; Nov 9-12, 2014; San Antonio, TX. Philadelphia (PA): AACR; Cancer Epidemiol Biomarkers Prev 2015;24(10 Suppl):Abstract nr PR09.


Molecular Cancer Research | 2014

Abstract A37: Complex crosstalk between MAPK signaling and energy metabolism in melanoma

Nathan J. Lanning; Pimiento Jose; Noel R. Monks; Paula J. Davidson; Andrew Borgman; Ting-Tung Chang; Anderson Peck; David Monsma; Chelsea A. Peterson; Mackeigan P. Jeff; Brian J. Nickoloff

The notoriously high glucose uptake by metastatic melanoma (MM) lesions is commonly exploited clinically using PET/CT imaging to evaluate disease progression and treatment. The purpose of this study was to use patient-derived xenograft (PDX) MM models and a PDX derived (MM8.1) cell line, to determine how driver oncogene mutations and drugs targeting the RAS-RAF-MEK signaling pathways impact glucose uptake, ATP production and mitochondrial plasticity of MM cells both in vivo and in vitro and the implications on drug resistance. MM BRAF V600E+ patients are routinely treated targeting either mutant BRAF kinase (vemurafenib), or the MAPK downstream signaling intermediate; MEK. To begin exploring links between oncogenic BRAF and metabolism, we first conducted differential gene expression (DGE) profiling of 30 different MM lesions (n=15 patients: pre- and post-engraftment). Focusing on PGC1α; a master mitochondrial regulator promoting oxidative phosphorylation (OXPHOS) revealed lower expression levels in BRAF V600E+ tumors compared to BRAF V600V+ tumors. Further analysis of a BRAF V600E+ MM PDX model revealed strong glucose uptake by PET/CT imaging at baseline. However, 48 hrs. after treatment with vemurafenib triggered growth arrest, accompanied by increased PGC1α; mRNA levels and significant reduction in glucose uptake without change in tumor volume. Upon acquisition of acquired vemurafenib resistance in the PDX model (day 70), rapidly growing tumors displayed reversal to high glucose uptake; which was again diminished by targeting MEK (PD0325901). DGE analysis of vehicle vs. 48 hrs. day 50 and day 70 vemurafenib treated tumors revealed oscillation in the expression of genes regulating glycolysis and oxidative phosphorylation (OXPHOS) that is reflected in the PET/CT imaging. To gain deeper insight, a cell line derived from a vehicle treated tumor, was examined using the Seahorse metabolic analyzer. Vehicle treated MM8.1 cells display high glycolytic capacity and predominantly glycolytic-derived ATP production, which is reflected by only minor inhibition of ATP production with oligomycin (an inhibitor of ATP synthase, a component of the OXHPOS system). Upon treatment with PLX4032, there was rapid reduction in glycolytic capacity along with a concurrent increase in maximum oxygen consumption capacity. PLX4032 treatment additionally enhanced MM8.1 sensitivity to oligomycin, but only transiently (24 hrs. pre-treatment), as later exposure times did not induce oligomycin sensitivity despite DGE changes to indicate otherwise. In conclusion, caution is warranted in evaluating drug responses in patients using PET/CT depending on duration of treatment. Cross-talk involving MAPK signaling and energy metabolism is a dynamic process, with more of a rheostat than switch-type properties, requiring further evaluation to identify therapeutic approaches exploiting metabolic events within MM cells. Citation Format: Nathan J. Lanning, Pimiento Jose, Noel R. Monks, Paula J. Davidson, Andrew S. Borgman, Ting-Tung Chang, Anderson S. Peck, David J. Monsma, Chelsea A. Peterson, Mackeigan P. Jeff, Brian J. Nickoloff. Complex crosstalk between MAPK signaling and energy metabolism in melanoma. [abstract]. In: Proceedings of the AACR Special Conference on RAS Oncogenes: From Biology to Therapy; Feb 24-27, 2014; Lake Buena Vista, FL. Philadelphia (PA): AACR; Mol Cancer Res 2014;12(12 Suppl):Abstract nr A37. doi: 10.1158/1557-3125.RASONC14-A37

Collaboration


Dive into the David Monsma's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge