Network


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

Hotspot


Dive into the research topics where Patrick J. Klein is active.

Publication


Featured researches published by Patrick J. Klein.


Journal of Clinical Oncology | 2012

Phase I Study of Folate Conjugate EC145 (Vintafolide) in Patients With Refractory Solid Tumors

Patricia LoRusso; Martin J. Edelman; Susan L. Bever; Karen M. Forman; MaryJo Pilat; Mary F. Quinn; Jing Li; Elisabeth I. Heath; Lisa Malburg; Patrick J. Klein; Christopher P. Leamon; Richard A. Messmann; Edward A. Sausville

PURPOSE EC145 (vintafolide), a conjugate of folic acid and the vinca alkaloid desacetylvinblastine hydrazide (DAVLBH), is a ligand for the folate receptor (FR), with activity against FR-positive tumor xenografts in vivo. This phase I study determined the maximum-tolerated dose (MTD) of EC145 administered as a bolus intravenous injection or 1-hour infusion in patients with refractory solid tumors. PATIENTS AND METHODS EC145 was administered as a bolus injection or 1-hour infusion on days 1, 3, and 5 and days 15, 17, and 19 of each 28-day cycle with dose escalation in cohorts of three to six patients until the MTD was identified. Plasma pharmacokinetics were determined on days 1 and 3 of the first cycle. RESULTS The MTD of EC145 was 2.5 mg when administered as either a bolus injection or 1-hour infusion. Constipation was the dose-limiting toxicity with both routes. Constipation, nausea, fatigue, and vomiting were the most commonly reported adverse events. One partial response to therapy was observed in a patient with metastatic ovarian cancer. CONCLUSION EC145 administered by bolus injection or as a 1-hour infusion at a dose of 2.5 mg on days 1, 3, and 5 and days 15, 17, and 19 of a 28-day cycle has an acceptable safety profile in patients with advanced cancer. On the basis of these findings, phase II studies of EC145 have been initiated in patients with advanced epithelial ovarian cancer and non-small-cell lung cancer.


Molecular Cancer Research | 2006

Parthenolide cooperates with NS398 to inhibit growth of human hepatocellular carcinoma cells through effects on apoptosis and G0-G1 cell cycle arrest.

Matthew Ralstin; Earl A. Gage; Michele T. Yip-Schneider; Patrick J. Klein; Eric A. Wiebke; C. Max Schmidt

Chemotherapy to date has not been effective in the treatment of human hepatocellular carcinoma. More effective treatment strategies may involve combinations of agents with activity against hepatocellular carcinoma. Parthenolide, a nuclear factor-κB (NF-κB) inhibitor, and NS398, a cyclooxygenase (COX)-2 inhibitor, have been shown to individually suppress the growth of hepatocellular carcinoma cells in vitro. To investigate their effects in combination, three human hepatocellular carcinoma lines (Hep3B, HepG2, and PLC) were treated with parthenolide and/or NS398. Parthenolide (0.1-10 μmol/L) and NS398 (1-100 μmol/L) each caused concentration-dependent growth inhibition in all cell lines. The addition of parthenolide to NS398 reduced the concentration of NS398 required to inhibit hepatocellular carcinoma growth. Because parthenolide and COX-2 inhibitors have been reported to influence NF-κB activity, the effects on this pathway were investigated. The combination of parthenolide/NS398 inhibited phosphorylation of the NF-κB-inhibitory protein IκBα and increased total IκBα levels. NF-κB DNA-binding and transcriptional activities were inhibited more by the combination than the single agents in Hep3B and HepG2 cells but not in PLC cells. The response of PLC cells to NS398 was augmented by p65 small interfering RNA to inhibit NF-κB p65 protein expression. The combination of parthenolide/NS398 increased apoptosis only in PLC cells, suggesting that the combination may decrease the apoptotic threshold in these cells. In Hep3B and HepG2 cells, combination treatment with NS398/parthenolide altered the cell cycle distribution resulting in more G0-G1 accumulation. Cyclin D1 levels were further decreased by combination treatment in all cell lines, correlating with the cell cycle alterations. Our results suggest that parthenolide may be effective in combination with COX-2 inhibitors for the treatment of hepatocellular carcinoma. (Mol Cancer Res 2006;4(6):387–400)


Journal of Pharmacology and Experimental Therapeutics | 2011

Reducing Undesirable Hepatic Clearance of a Tumor-Targeted Vinca Alkaloid via Novel Saccharopeptidic Modifications

Christopher P. Leamon; Joseph A. Reddy; Patrick J. Klein; Iontcho Radoslavov Vlahov; Ryan Dorton; Alicia Bloomfield; Melissa Nelson; Elaine Westrick; Nikki Parker; Kristen Bruna; Marilynn Vetzel; Mark A Gehrke; Jeffrey S. Nicoson; Richard A. Messmann; Patricia LoRusso; Edward A. Sausville

During a phase I trial of EC145 (a folate-targeted vinca alkaloid conjugate), constipation was identified as the dose-limiting toxicity, probably from a nonfolate receptor-related liver clearance process capable of releasing unconjugated vinca alkaloid from EC145 and shuttling it to the bile. Here, we report on the selective placement of novel carbohydrate segments (1-amino-1-deoxy-glucitolyl-γ-glutamate) spaced in-between the folate and vinca alkaloid moieties of EC145, which yielded a new agent (EC0489) that is equipotent but less toxic than EC145. Whereas both compounds could cure tumor-bearing mice reproducibly, EC0489 differed from EC145 with i) a shorter elimination half-life, ii) approximately 70% decrease in bile clearance, iii) a 4-fold increase in urinary excretion, and iv) improved tolerability in rodents. This combination of improvements justified the clinical evaluation of EC0489 where currently administered dose levels have exceeded the maximal tolerated dose of EC145 by approximately 70%, thereby reflecting the translational benefits to this new approach.


Journal of Pharmacology and Experimental Therapeutics | 2009

Resistance to Mitogen-Activated Protein Kinase Kinase (MEK) Inhibitors Correlates with Up-Regulation of the MEK/Extracellular Signal-Regulated Kinase Pathway in Hepatocellular Carcinoma Cells

Michele T. Yip-Schneider; Patrick J. Klein; Sabrina C. Wentz; Amer Zeni; Alex Menze; C. Max Schmidt

The extracellular signal-regulated (ERK), mitogen-activated protein kinase (p42/p44 MAPK) pathway is up-regulated in hepatocellular carcinoma (HCC). Molecular targeting of this critical mitogenic pathway may have therapeutic potential for the treatment of HCC; however, chemoresistance to long-term therapy may develop. In the present study, we employed small-molecule MAPK kinase (MEK) inhibitors, including U0126 [1,4-diamino-2,3-dicyano-1,4-bis(2-aminophynyltio)butadiene] and PD184161 (Neoplasia 8:1–8, 2006), in HepG2 and Hep3B human HCC cell lines to identify potential mechanism(s) of resistance. U0126 dose-dependently suppressed ERK phosphorylation at both 1- and 24-h time points in HepG2 cells, previously shown to be sensitive to growth inhibition by U0126. In contrast, ERK phosphorylation was only decreased at the 1-h time point but not at 24 h in the more resistant Hep3B cells. It is interesting that the lack of prolonged phospho-ERK suppression was associated with MEK hyperphosphorylation in Hep3B cells. Several MEK/ERK pathway intermediates were up-regulated in Hep3B cells; furthermore, transfection of Raf-1 small interfering RNA to suppress MEK/ERK pathway activation sensitized Hep3B cells to U0126. MEK inhibitor resistance was independent of p53 or hepatitis Bx protein status. Finally, we showed that combining two chemically distinct MEK inhibitors enhanced growth inhibition and apoptosis compared with the single agents. Taken together, these results suggest that up-regulated expression or activity of the MEK/ERK pathway contributes to MEK inhibitor resistance in HCC cells. Our findings also provide preclinical evidence suggesting that the status of the MEK/ERK pathway in patients may predict response to MEK/ERK-targeted therapeutics.


Hepatology | 2010

Targeting mitogen-activated protein kinase kinase with the inhibitor PD0325901 decreases hepatocellular carcinoma growth in vitro and in mouse model systems†

Matthew Hennig; Michele T. Yip-Schneider; Sabrina C. Wentz; Huangbing Wu; S. K. Hekmatyar; Patrick J. Klein; Navin Bansal; C. Max Schmidt

Hepatocellular carcinoma (HCC) is a common cause of death from solid organ malignancy worldwide. Extracellular signal‐regulated/mitogen‐activated protein kinase kinase (MEK) signaling is a critical growth regulatory pathway in HCC. Targeting MEK with a novel small molecule inhibitor, PD0325901, may inhibit HCC tumorigenesis. PD0325901 (0.01‐100 nM) inhibited growth and MEK activity in vitro in immortalized murine transforming growth factor alpha (TGF‐α) transgenic hepatocyte (TAMH) cells, derived from the livers of TGF‐α transgenic mice. Treatment of athymic mice bearing TAMH flank tumors with vehicle or PD0325901 (20 mg/kg) revealed a significant reduction of MEK activity ex vivo 24 hours after a single PD0325901 dose. The growth rate of TAMH flank tumors over 16 days was reduced threefold in the treatment arm (1113 ± 269% versus 3077 ± 483%, P < 0.01). PD0325901 exhibited similar inhibitory effects in HepG2 and Hep3B human HCC cells in vitro and in Hep3B flank tumors in vivo. To confirm this in a developmental model, MT‐42 (CD‐1) TGF‐α mice were treated with vehicle or PD0325901 (20 mg/kg) for 5 weeks. Gross HCC was detected in 47% and 13.3% of the control and treatment mice, respectively. Tumor growth suppression by PD0325901 relative to vehicle was also shown by magnetic resonance imaging. These studies provide compelling preclinical evidence that targeting MEK in human clinical trials may be promising for the treatment of HCC. (HEPATOLOGY 2010.)


Journal of Surgical Research | 2009

Ethanol-TGFα-MEK Signaling Promotes Growth of Human Hepatocellular Carcinoma

Matthew Hennig; Michele T. Yip-Schneider; Patrick J. Klein; Sabrina C. Wentz; Jesus M. Matos; Courtney J. Doyle; Jennifer N. Choi; Huangbing Wu; Amanda O'Mara; Alex Menze; Stephen Noble; Iain H. McKillop; C. Max Schmidt

BACKGROUND Chronic ethanol intake is a significant risk factor for the development of cirrhosis and hepatocellular carcinoma (HCC). The effects of ethanol on extracellular signal-regulated kinase (ERK) activation, transforming growth factor alpha (TGF-alpha), and HCC growth were examined in this study. METHODS HepG2, SKHep, Hep3B human HCC cells, or normal human hepatocytes were treated with ethanol (0-100 mM), exogenous TGF-alpha, TGF-alpha neutralization antibody or the MEK inhibitor U0126. TGF-alpha levels were quantified by ELISA. Growth was determined by trypan blue-excluded cell counts. Cell cycle phase distribution was determined by flow cytometry. Protein expression was determined by Western blot. RESULTS Ethanol treatment (10-40 mM) increased ERK activation in HepG2 and SKHep HCC cells but not in Hep3B or human hepatocyte cells. Growth increased in HepG2 (174 +/- 29%, P < 0.05) and SKHep (149 +/- 12%, P < 0.05) cells in response to ethanol treatment. Correspondingly, ethanol increased S phase distribution in these cells. U0126 suppressed ethanol-induced growth increases. Ethanol treatment for 24 h also raised TGF-alpha levels in HepG2 cells (118%-198%) and SKHep cells (112%-177%). Exogenous administration of recombinant TGF-alpha mimicked the ethanol-induced growth in HepG2 and SKHep cells; TGF-alpha neutralization antibody effectively abrogated this effect. The TGF-a neutralization antibody also prevented ERK activation by ethanol in HepG2 cells. CONCLUSIONS These data demonstrate that clinically relevant doses of ethanol stimulate ERK-dependent proliferation of HCC cells. Ethanol up-regulates TGF-alpha levels in HCC cells and enhances growth through cell cycles changes, which appear to be mediated through TGF-alpha-MEK-ERK signaling. Ethanol-MEK signaling in normal hepatocytes is absent, suggesting that ethanol promotion of HCC growth may in part depend upon the acquisition of cancer-specific signaling by hepatocytes.


Cancer Research | 2017

Abstract 2057: Evaluation of anti-tumor efficacy of EC1456 in low-passage and pre-treated patient-derived xenograft models of triple-negative breast cancer

Yingjuan Lu; Nikki Parker; Haiyan Chu; Michael R. Pugh; Satish Rao; Patrick J. Klein; Michael F. Ritchie; Lonnie Myer; Jennifer Jaskowiak; Christopher P. Leamon

Triple negative breast cancer (TNBC) patients are insensitive to hormonal or anti-HER2 therapy and have a higher recurrence rate among all breast cancer subtypes. There is a lack of common therapeutic targets in TNBC due to its six distinct molecular characteristics. Recently, ~50% of TNBC cases were found to express the folate receptor alpha (FRα) on tumor cells. FRα is a GPI-anchored membrane glycoprotein capable of bringing folate-targeted small-molecule drug conjugates (SMDCs) inside the cell. EC1456 is a folic acid-tubulysin B hydrazide (TubBH) SMDC that specifically binds to the membrane FRα and is internalized by endocytosis. While encapsulated within the early endosome, EC1456 releases TubBH into the cytosol where it inhibits the polymerization of tubulin into microtubules, thus blocking spindle formation to arrest cells in metaphase which ultimately induces apoptosis. EC1456 is currently under Phase 1 clinical investigation in patients with common solid tumors [IND# 118,859]. The purpose of this study is to evaluate EC1456 activity in Champions TumorGraftTM TNBC patient-derived xenograft (PDX) models to help guide our drug development strategies. These PDX models were derived from patients who were treated with multiple lines of standard-of-care agents. A total of six low-passage, FR-positive TNBC models were tested against two different treatment regimens of EC1456 (once or twice a week for 2 weeks only). Plasma and tumor drug concentrations were quantified by LC-MS/MS using satellite study animals. The tumor-bearing animals were monitored for up to 60 days to assess both short-term (i.e. % TGI) and long-term (%PR, CR, TFS) anti-tumor responses. Using a stringent efficacy criteria (≥60% CR/TFS), 3 of the 6 TNBC models were found highly sensitive to EC1456 and 3 were found resistant. To identify potential gene signatures of EC1456 response, bioinformatics analysis was performed using existing RNA-seq data and compared across a broad panel of TumorGraftTM TNBC models, regardless of FR expression status. Specific biomarkers of interest were further analyzed by qRT-PCR using control tumors from the current study. Together, our analysis revealed potential resistance mechanisms associated with microtubule dynamics as well as a cancer cell’s ability to undergo apoptosis. Citation Format: Yingjuan Lu, Nikki L. Parker, Haiyan Chu, Michael R. Pugh, Satish I. Rao, Patrick J. Klein, Michael F. Ritchie, Lonnie D. Myer, Jennifer Jaskowiak, Christopher P. Leamon. Evaluation of anti-tumor efficacy of EC1456 in low-passage and pre-treated patient-derived xenograft models of triple-negative breast cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2057. doi:10.1158/1538-7445.AM2017-2057


Cancer Research | 2012

Abstract LB-5: Folate-targeted therapy for invasive urinary bladder cancer

Deepika Dhawan; José A. Ramos-Vara; James F. Naughton; Liang Cheng; Phillip S. Low; Ryan Rothenbuhler; Christopher P. Leamon; Nikki Parker; Patrick J. Klein; Iontcho Radoslavov Vlahov; Lindsey M. Fourez; Linda M. Murphy; Deborah W. Knapp

Introduction: Invasive transitional cell carcinoma of the urinary bladder (InvTCC) kills >14,000 people yearly in the US. Folate receptors (FRs, especially FRα) are being targeted for imaging and therapy across multiple cancer types. This work was performed to define FR expression and uptake in InvTCC, with comparison to normal bladder; and to determine the antitumor activity and toxicity of folate-targeted vinblastine in dogs with naturally-occurring InvTCC where the cancer closely mimics human InvTCC. Canine InvTCC metastasizes frequently, and only 35% of dogs have remission with chemotherapy. Methods: Immunohistochemistry (IHC, mab343, PU17, FRβmab) was performed in InvTCC, bladder tissues adjacent to InvTCC in humans, and in bladder from normal dogs. Folate uptake was determined ex vivo by folate binding assay, and in vivo in dogs with InvTCC by nuclear scintigraphy ( 99m Tc based FR targeted imaging agent). A dose escalation study of folate-targeted vinblastine (EC0905, IV weekly) was performed in dogs with biopsy-confirmed, FR-positive, measurable InvTCC. Toxicity (VCOG criteria) was assessed by CBCs, serum chemistry profiles, urinalyses, physical exams, and dog owner observations. The MTD was defined as the highest dose at which 0 of 6 dogs had grade 4 toxicity and ≤1 of 6 dogs had grade 3 toxicity. Tumor response was assessed by ultrasonography (with detailed bladder mapping) and radiography. Results: FR expression (IHC, PU17) was noted in the majority of canine samples including 56 of 74 (76%) primary tumors, 7 of 12 (58%) nodal metastases, and 10 of 21 (48%) lung metastases, as well as in normal urothelium. Typically, >50% of tumor cells were positive. FR expression was cytoplasmic in 18, membrane in 6, and in both sites in 32 InvTCC cases. Scintigraphy (n= 12 dogs) revealed folate uptake in primary and metastatic lesions. The MTD of EC0905 in dogs was 0.25 mg/kg IV weekly. At doses above the MTD, gastrointestinal upset and myelosuppression occurred. No urologic toxicity was noted. Tumor responses included partial remission (≥50% reduction in tumor volume) in 5 dogs and stable disease ( Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr LB-5. doi:1538-7445.AM2012-LB-5


Neoplasia | 2006

The Effects of a Novel MEK Inhibitor PD184161 on MEK-ERK Signaling and Growth in Human Liver Cancer

Patrick J. Klein; C. Max Schmidt; Chad A. Wiesenauer; Jennifer N. Choi; Earl A. Gage; Michele T. Yip-Schneider; Eric A. Wiebke; Yufang Wang; Charles Omer; Judith Sebolt-Leopold


Journal of Gastrointestinal Surgery | 2008

Targeting MEK is effective chemoprevention of hepatocellular carcinoma in TGF-α-transgenic mice

Sabrina C. Wentz; Huangbing Wu; Michele T. Yip-Schneider; Matthew Hennig; Patrick J. Klein; Judith Sebolt-Leopold; C. Max Schmidt

Collaboration


Dive into the Patrick J. Klein'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
Researchain Logo
Decentralizing Knowledge