Cancer Letters

Cancer Letters

Volume 432, 28 September 2018, Pages 251-259
Cancer Letters

Original Articles
Oral recombinant methioninase (o-rMETase) is superior to injectable rMETase and overcomes acquired gemcitabine resistance in pancreatic cancer

https://doi.org/10.1016/j.canlet.2018.06.016Get rights and content

Highlights

  • Evaluated the efficacy of Oral methioninase (o-rMETase) on a pancreatic cancer PDOX model.

  • o-rMETase is highly effective against the pancreatic cancer PDOX.

  • o-rMETase is potentially safer than intra-peritoneally-methioninase.

  • o-rMETase overcomes acquired gemcitabine resistance in pancreatic cancer.

Abstract

Recombinant methioninase (rMETase) was previously administered as an injectable drug to target methionine dependence of cancer. Recently, we observed that rMETase could be administered orally (o-rMETase) in a patient-derived orthotopic xenograft (PDOX) mouse model of melanoma. Here, we determined the efficacy of o-rMETase on a pancreatic cancer PDOX model. Forty pancreatic cancer PDOX mouse models were randomized into four groups of 10 mice each. o-rMETase was significantly more effective than i.p.-rMETase, but the combination of both was significantly more effective than either alone. Acquired gemcitabine resistance is a major factor in the recalcitrance of pancreatic cancer. We tested a human pancreatic cancer cell line, which has acquired >100-fold GEM-resistance (PK-9R) than its parental cell line PK-9. In contrast to GEM, both cell lines were very sensitive to rMETase. In orthotopic nude mouse models of PK-9 and PK-9R, GEM inhibited tumor growth in PK-9 but not PK-9R. In contrast, o-rMETase could inhibit both tumors. The combination of GEM + o-rMETase could regress the PK-9 tumor and inhibit PK-9R tumor growth. The present study shows that o-rMETase is effective and overcomes acquired GEM resistance in pancreatic cancer and demonstrates the clinical potential of this strategy.

Introduction

The elevated methionine (MET) requirement of cancer cells to proliferate is termed MET dependence and may be the only known general metabolic defect in cancer [1,2]. Cancer cells utilize excess methionine for aberrant transmethylation reactions [3], a metabolic reprogramming in cancer known as the “Hoffman Effect” and analogous to the Warburg effect of over-utilization of glucose in cancer [2]. In head-to-head comparisons in PET imaging, [11C] MET-PET shows a more stronger signal than [18F] fluorodeoxyglucose [4], suggesting that the Hoffman effect is more extensive in cancer than the Warburg effect. Targeting MET by recombinant methioninase (rMETase) can arrest the growth of cancer cells in vitro and in vivo [[1], [2], [3], [4]].

METase has been used as a therapeutic strategy for multiple type of cancers [[4], [5], [6], [7], [8], [9], [10], [11]]. Song et al. [12] reported that MET starvation therapy using a MET-free diet or total parenteral nutrition (TPN) extends the survival time of high-stage gastric carcinoma patients.

Previously rMETase was administrated by intra-peritoneal injection (ip-rMETase) and was effective against patient-derived orthotopic xenograft (PDOX) models of recalcitrant cancer [[13], [14], [15], [16], [17]]. Recently, we compared ip-rMETase and oral rMETase (o-rMETase) for efficacy on the melanoma PDOX [13]. o-rMETase was significantly more effective than ip-rMETase [13], indicating the potential widespread use of rMETase for acute and chronic cancer treatment.

Pancreatic cancer is the fourth leading causes of cancer-related deaths in the Western world because of low-response to radio- and chemo-therapies. The American Cancer Society estimates that in 2018, about 55,440 new cased will be diagnosed with pancreatic cancer and about 44,330 people will die of this disease in the USA alone [18]. Despite extensive research to improve detection techniques, surgical methods and developing better chemotherapy drugs, the prognosis of pancreatic cancer patients remains poor.

Gemcitabine (GEM) is the standard first line treatment for patients with advanced pancreatic cancer; however, it is ineffective because of acquired resistance [19,20]. Acquired GEM resistance is a major factor in the recalcitrance of pancreatic cancer. Various genetic and/or epigenetic changes have been proposed that causes GEM resistance [[20], [21], [22], [23], [24], [25]]; but how to overcome this resistance remains unclear.

We recently reported that intra-peritoneal rMETase (i.p.-rMETase) combined with GEM could regress a partially GEM-resistant pancreatic cancer PDOX [26]. In the present study, we compared the efficacy of o-rMETase and i.p.-rMETase in a pancreatic-cancer PDOX. Acquired GEM resistance is a major factor in the recalcitrance of pancreatic cancer. In addition, we describe a human pancreatic cancer cell line which acquired greater than 100-fold GEM resistance. We evaluated the efficacy of o-rMETase and o-rMETase in combination with GEM against the GEM-resistant human PK-9R pancreatic cancer cell line in vitro and orthotopic mouse models compared to its parental GEM-sensitive line PK-9. We found that o-rMETase can overcome GEM resistance.

Section snippets

Mice

Athymic nu/nu nude mice (AntiCancer Inc., San Diego, CA), 4–6 weeks old, were used in this study. All experimental protocols and data collection were as previously described [[13], [14], [15], [16], [17]]. All mouse surgical procedures and imaging were performed with the animals anesthetized by subcutaneous injection of a ketamine mixture were as previously described [[13], [14], [15], [16], [17]]. The response of animals during surgery was monitored to ensure adequate depth of anesthesia. The

Efficacy of rMETase on a PDOX model of pancreatic cancer

To test which rMETase (o-rMETase or i.p.-rMETase) can effectively inhibit a PDOX model of pancreatic cancer, we treated mice with i.p.-rMETase, or o-rMETase alone or the combinations of o-rMETase + i.p.-rMETase twice a day for 14 consecutive days and compared them with untreated control group. All treatments significantly inhibited tumor growth compared to the untreated control (i.p.-rMETase: p < 0.0001; o-rMETase: p < 0.0001; o-rMETase + i.p.-rMETase: p < 0.0001); on day 14 after initiation of

Discussion

In the present study using a PDOX model, we show that o-rMETase is more effective than injectable rMETase for pancreatic cancer. In addition, we show that o-rMETase can overcome acquired GEM resistance in pancreatic cancer and demonstrates the clinical potential of this strategy. We have previously shown the effect of injectable rMETase on tumor histology [[14], [15], [16]]. Detailed histological effects of o-rMETase will be part of future studies.

We have developed PDOX mouse models of cancer

Conflicts of interest

The authors declare that they have no competing interests.

Acknowledgements

This paper is dedicated to the memory of A. R. Moossa, M.D., Sun Lee, M.D. and Shigeo Yagi, Ph.D.

References (101)

  • R.M. Hoffman

    Development of recombinant methioninase to target the general cancer-specific metabolic defect of methionine dependence: a 40-year odyssey

    Expet Opin. Biol. Ther.

    (2015)
  • R.M. Hoffman

    The wayward methyl group and the cascade to cancer

    Cell Cycle

    (2017)
  • P.H. Stern et al.

    Elevated overall rates of transmethylation in cell lines from diverse human tumors

    In Vitro – Rapid Commun. Cell Biol.

    (1984)
  • W. Xu et al.

    The performance of 11C-Methionine PET in the differential diagnosis of glioma recurrence

    Oncotarget

    (2017)
  • L. Xin et al.

    Stealth cationic liposomes modified with anti-CAGE single-chain fragment variable deliver recombinant methioninase for gastric carcinoma therapy

    J. Nanosci. Nanotechnol.

    (2013)
  • L. Xin et al.

    Evaluation of METase-pemetrexed-loaded PEG-PLGA nanoparticles modified with anti-CD133-scFV for treatment of gastric carcinoma

    Biosci. Rep.

    (2018)
  • Y. Tan et al.

    Broad selective efficacy of recombinant methioninase and polyethylene glycol-modified recombinant methioninase on cancer cells in vitro

    Anticancer Res.

    (2010)
  • W. Kreis et al.

    Biological effects of enzymatic deprivation of L-methionine in cell culture and an experimental tumor

    Cancer Res.

    (1973)
  • S. Yano et al.

    Tumor-specific cell-cycle decoy by Salmonella typhimurium A1-R combined with tumor-selective cell-cycle trap by methioninase overcome tumor intrinsic chemoresistance as visualized by FUCCI imaging

    Cell Cycle

    (2016)
  • S. Yano et al.

    Selective methioninase-induced trap of cancer cells in S/G2 phase visualized by FUCCI imaging confers chemosensitivity

    Oncotarget

    (2014)
  • S. Yano et al.

    Tumor-targeting Salmonella typhimurium A1-R decoys quiescent cancer cells to cycle as visualized by FUCCI imaging and become sensitive to chemotherapy

    Cell Cycle

    (2011)
  • Y.Q. Song et al.

    Cost-effective comparison between early enteral nutrition and total parenteral nutrition on elder postoperative gastric cancer patients

    Chin. J. Gen. Surg.

    (2004)
  • K. Kawaguchi et al.

    Combination treatment with recombinant methioninase enables temozolomide to arrest a BRAF V600E melanoma growth in a patient-derived orthotopic xenograft

    Oncotarget

    (2017)
  • K. Kawaguchi et al.

    Recombinant methioninase (rMETase) is an effective therapeutic for BRAF-V600E-negative as well as -positive melanoma in patient-derived orthotopic xenograft (PDOX) mouse models

    Oncotarget

    (2018)
  • K. Igarashi et al.

    Growth of a doxorubicin-resistant undifferentiated spindle-cell sarcoma PDOX is arrested by metabolic targeting with recombinant methioninase

    J. Cell. Biochem.

    (2018)
  • K. Igarashi et al.

    Tumor-targeting Salmonella typhimurium A1-R combined with recombinant methioninase and cisplatinum eradicates an osteosarcoma cisplatinum-resistant lung metastasis in a patient-derived orthotopic xenograft (PDOX) mouse model: decoy, trap and kill chemotherapy moves toward the clinic

    Cell Cycle

    (2018)
  • American Cancer Society Key Statistics for Pancreatic Cancer (accessed on May 1. 2018)....
  • H.A. Burris et al.

    Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial

    J. Clin. Oncol.

    (1997)
  • H. Yoneyama et al.

    Acquired resistance to gemcitabine and cross-resistance in human pancreatic cancer clones

    Anti Canc. Drugs

    (2015)
  • J. Garcia-Manteiga et al.

    Nucleoside transporter profiles in human pancreatic cancer cells: role of hCNT1 in 2′,2′-difluorodeoxycytidine-induced cytotoxicity

    Clin. Canc. Res.

    (2003)
  • E. Giovannetti et al.

    Transcription analysis of human equilibrative nucleoside transporter-1 predicts survival in pancreas cancer patients treated with gemcitabine

    Canc. Res.

    (2006)
  • Y. Nakano et al.

    Gemcitabine chemoresistance and molecular markers associated with gemcitabine transport and metabolism in human pancreatic cancer cells

    Br. J. Canc.

    (2007)
  • S.S. Liau et al.

    HMGA1 is a molecular determinant of chemoresistance to gemcitabine in pancreatic adenocarcinoma

    Clin. Canc. Res.

    (2008)
  • K. Kawaguchi et al.

    Targeting altered cancer methionine metabolism with recombinant methioninase (rMETase) overcomes partial gemcitabine-resistance and regresses a patient-derived orthotopic xenograft (PDOX) nude mouse model of pancreatic cancer

    Cell Cycle

    (2018)
  • K. Kawaguchi et al.

    Vemurafenib-resistant BRAF-V600E mutated melanoma is regressed by MEK targeting drug trametinib, but not cobimetinib in a patient-derived orthotopic xenograft (PDOX) mouse model

    Oncotarget

    (2016)
  • K. Kawaguchi et al.

    Tumor-targeting Salmonella typhimurium A1-R combined with temozolomide regresses malignant melanoma with a BRAF-V600 mutation in a patient-derived orthotopic xenograft (PDOX) model

    Oncotarget

    (2016)
  • K. Kawaguchi et al.

    Tumor-targeting Salmonella typhimurium A1-R sensitizes melanoma with a BRAF-V600E mutation to vemurafenib in a patient-derived orthotopic xenograft (PDOX) nude mouse model

    J. Cell. Biochem.

    (2017)
  • A. Suetsugu et al.

    Multi-color palette of fluorescent proteins for imaging the tumor microenvironment of orthotopic tumorgraft mouse models of clinical pancreatic cancer specimens

    J. Cell. Biochem.

    (2012)
  • A. Suetsugu et al.

    Imageable fluorescent metastasis resulting in transgenic GFP mice orthotopically implanted with human-patient primary pancreatic cancer specimens

    Anticancer Res.

    (2012)
  • A. Suetsugu et al.

    Non-invasive fluorescent-protein imaging of orthotopic pancreatic-cancer-patient tumorgraft progression in nude mice

    Anticancer Res.

    (2012)
  • X. Fu et al.

    A metastatic nude-mouse model of human pancreatic cancer constructed orthotopically from histologically intact patient specimens

    Proc. Natl. Acad. Sci. U. S. A.

    (1992)
  • R.M. Hoffman
  • X. Fu et al.

    A metastatic-orthotopic transplant nude-mouse model of human patient breast cancer

    Anticancer Res.

    (1993)
  • X. Fu et al.

    Human ovarian carcinoma metastatic models constructed in nude mice by orthotopic transplantation of histologically-intact patient specimens

    Anticancer Res.

    (1993)
  • X. Wang et al.

    A new patient-like metastatic model of human lung cancer constructed orthotopically with intact tissue via thoracotomy in immunodeficient mice

    Int. J. Canc.

    (1992)
  • Y. Hiroshima et al.

    Establishment of a patient-derived orthotopic xenograph (PDOX) model of HER-2-positive cervical cancer expressing the clinical metastatic pattern

    PLoS One

    (2015)
  • X. Fu et al.

    Models of human metastatic colon cancer in nude mice orthotopically constructed by using histologically intact patient specimens

    Proc. Natl. Acad. Sci. U. S. A.

    (1991)
  • C.A. Metildi et al.

    Fluorescently-labeled chimeric anti-CEA antibody improves detection and resection of human colon cancer in a patient-derived orthotopic xenograft (PDOX) nude mouse model

    J. Surg. Oncol.

    (2014)
  • Y. Hiroshima et al.

    Successful fluorescence-guided surgery on human colon cancer patient-derived orthotopic xenograft mouse models using a fluorophore-conjugated anti-CEA antibody and a portable imaging system

    J. Laparoendosc. Adv. Surg. Tech.

    (2014)
  • K. Kawaguchi et al.

    Intra-tumor L-methionine level highly correlates with tumor size in both pancreatic cancer and melanoma patient-derived orthotopic xenograft (PDOX) nude-mouse models

    Oncotarget

    (2018)
  • Cited by (0)

    View full text