Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • Anticancer Research
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • In Vivo
    • Anticancer Research
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
In Vivo
  • Other Publications
    • In Vivo
    • Anticancer Research
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
In Vivo

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • Anticancer Research
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
  • About Us
    • General Policy
    • Contact
  • Visit iiar on Facebook
  • Follow us on Linkedin
Research ArticleExperimental Studies
Open Access

Chloroquine Combined With Rapamycin Arrests Tumor Growth in a Patient-derived Orthotopic Xenograft (PDOX) Mouse Model of Dedifferentiated Liposarcoma

NORIYUKI MASAKI, YUSUKE AOKI, YUTARO KUBOTA, KOYA OBARA, JUN MIYAZAKI and ROBERT M. HOFFMAN
In Vivo November 2022, 36 (6) 2630-2637; DOI: https://doi.org/10.21873/invivo.12997
NORIYUKI MASAKI
1AntiCancer Inc, San Diego, CA, U.S.A.;
2Graduate School of Medicine, International University of Health and Welfare, Tokyo, Japan;
3Department of Surgery, University of California, San Diego, CA, U.S.A.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
YUSUKE AOKI
1AntiCancer Inc, San Diego, CA, U.S.A.;
3Department of Surgery, University of California, San Diego, CA, U.S.A.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
YUTARO KUBOTA
1AntiCancer Inc, San Diego, CA, U.S.A.;
3Department of Surgery, University of California, San Diego, CA, U.S.A.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
KOYA OBARA
1AntiCancer Inc, San Diego, CA, U.S.A.;
3Department of Surgery, University of California, San Diego, CA, U.S.A.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
JUN MIYAZAKI
4Department of Urology, School of Medicine, International University of Health and Welfare, Narita, Japan
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
ROBERT M. HOFFMAN
1AntiCancer Inc, San Diego, CA, U.S.A.;
3Department of Surgery, University of California, San Diego, CA, U.S.A.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: all@anticancer.com
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background/Aim: Dedifferentiated liposarcoma (DDLS) is a type of soft-tissue sarcoma with a poor prognosis due to distant metastasis and resistance to chemotherapy. The antimalarial drug chloroquine (CQ) can induce apoptosis in cancer cells. CQ in combination with rapamycin (RAPA), an mTOR inhibitor, has shown efficacy on osteosarcoma and other types of cancer. In the present study the efficacy of RAPA combined with CQ on the treatment of a DDLS patient-derived orthotopic xenograft (PDOX) model was investigated. Materials and Methods: A patient-derived DDLS was transplanted into the left retroperitoneum of nude mice to establish a DDLS PDOX nude-mouse model. The mice were randomly divided as follows: untreated control group; CQ group; RAPA group; combined CQ and RAPA group (n=7 for all groups). During the treatment period, tumor volume was measured every 3-4 days with calipers. After 2 weeks treatment, the mice were sacrificed, and H&E staining was performed for histological evaluation. The TUNEL assay was performed to detect apoptosis. Results: The combination of CQ and RAPA arrested tumor growth in the DDLS PDOX compared to the untreated control (p=0.009) and was significantly more effective than RAPA alone (p=0.009). RAPA alone slowed tumor growth, but the difference was not statistically significant (p>0.05). CQ was not active alone (p>0.05). The number of apoptotic TUNEL-positive cells was significantly higher in the CQ plus RAPA group than in the other groups (p=0.02). Conclusion: Combination therapy with CQ and RAPA arrested tumor growth in a DDLS PDOX model by inducing apoptosis.

Key Words:
  • Dedifferentiated liposarcoma
  • PDOX
  • patient-derived orthotopic xenograft
  • combination therapy
  • mTOR inhibitors
  • rapamycin
  • chloroquine
  • apoptosis

Dedifferentiated liposarcoma (DDLS) has a poor prognosis due to distant metastasis and resistance to chemotherapy (1, 2). For patients with recurrent or metastatic dedifferentiated liposarcoma, ifosfamide and doxorubicin are first-line treatment (3-7), but with limited efficacy (1).

Our laboratory established the technique of surgical orthotopic implantation in 1988, pioneering the patient-derived orthotopic xenograft (PDOX) mouse model (8, 9). We have shown that the PDOX mouse model, unlike the subcutaneous PDX mouse model, retains the metastatic potential of the original tumor after transplantation in nude mice (9, 10).

Rapamycin (RAPA), a mammalian target of rapamycin (mTOR) inhibitor, is used in transplant medicine as an immunosuppressive agent (11), as well as for treatment of renal cell carcinoma (12). mTOR plays a central role in the regulation of various cellular functions such as survival and proliferation (13). We have previously demonstrated the anticancer effects of mTOR inhibitors in combination with multiple agents in PDOX mouse models of osteosarcoma (14, 15).

Chloroquine (CQ) is an antimalarial drug developed in Germany in 1934 (16). CQ is an autophagy inhibitor and can induce apoptosis in cancer cells (17). Previous studies have demonstrated the efficacy of CQ in combination with cancer-chemotherapy drugs in metastatic prostate cancer cell lines and hepatocarcinoma cell lines via increased apoptosis (18, 19). The combination of CQ and the mTOR inhibitor temsirolimus has shown efficacy in solid tumors and melanoma, in a Phase I clinical trial (20).

In the present study, we established a PDOX mouse model of DDLS and investigated the efficacy of combination treatment with CQ and RAPA.

Materials and Methods

Animals. Athymic (nu/nu) nude male mice, 4-6 weeks old, (AntiCancer, Inc., San Diego, CA, USA) were used under an AntiCancer, Inc. Institutional Animal Care and Use Committee (IACUC) protocol. This was specifically approved for the present study, which followed the principles and procedures outlined in the National Institutes of Health Guide for the Care and Use of Animals, Assurance No. A3873-1.

Patient-derived tumor. The DDLS surgical specimen was from a primary retroperitoneal DDLS in a male in his 80s. The patient previously underwent surgical resection. The patient’s tumor was provided as a discarded pathology specimen at the Department of Surgery, University of California, Los Angeles, CA, USA (UCLA). All experiments were performed in accordance with the Declaration of Helsinki and regulations on human research. Informed consent was obtained from the patient as part of a UCLA Institutional Review board approval protocol to obtain the tumor specimen (IRB #10-001857).

Establishment of a DDLS PDOX nude-mouse model. The tumor was minced to approximately 40 mm3 in size (Figure 1). All surgical procedures were performed under anesthesia induced by a ketamine mixture. After anesthesia, a 1.5 cm incision was made in the left retroperitoneum (Figure 2A), and a 40 mm3 tumor was implanted on the retroperitoneum in the back of the kidney (Figure 2B). The wound was closed with 5-0 nylon sutures (Figure 2C). The tumor grew to 50-100 mm3 in size within 10 days of implantation and could be measured through the skin (Figure 2D).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Minced fragments of dedifferentiated liposarcoma tumor tissue and surrounding normal tissue prepared for orthotopic implantation.

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Establishment of a dedifferentiated liposarcoma patient-derived orthotopic xenograft model. (A) Left retroperitoneal incision of approx. 1.5 cm. White arrow indicates left kidney. (B) The tumor was sutured to the fatty tissue of the retroperitoneum in the back of the kidney with an 8-0 nylon suture. White arrow indicates the implanted tumor fragment. (C) The wound was closed with 5-0 nylon sutures. (D) White arrow indicates tumor grown to approximately 70 mm3, 10 days after transplantation.

Reagents. Rapamycin (HY-10219) (MedChemExpress, Monmouth Junction, NJ, USA) was dissolved in phosphate-buffered saline (PBS) with 4.8% Tween 80, 4.8% polyethylene glycol 400 and 4% ethanol. Chloroquine diphosphate (C6628) (Sigma-Aldrich, St. Louis, MO, USA) was dissolved in PBS.

Treatment schedule. Ten days after orthotopic transplantation, treatment was initiated when tumor volume reached 50-100 mm3 and continued for 15 days. The DDLS-PDOX mouse model was randomly assigned to four groups of 7 mice per group as follows: untreated control; CQ [100.0 mg/kg/day, intraperitoneal (i.p.) injection]; RAPA (1.0 mg/kg/day, i.p. injection); combination of CQ (100.0 mg/kg/day, i.p. injection) and RAPA (1.0 mg/kg/day, i.p. injection) (Figure 3).

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Treatment scheme. CQ, Chloroquine; RAPA, rapamycin.

The short and long axes of the tumors were measured using calipers, and the mice were weighed once a week. Tumor volume was determined as (short-axis diameter)2×long-axis diameter×0.5. All mice were sacrificed on day 15 after administration. Tumors were then removed for histological evaluation.

Hematoxylin and eosin staining. Specimens were prepared and stained according to the standard hematoxylin-eosin protocol for histo-pathological evaluation (21).

Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. Apoptosis was assessed using the One-step TUNEL In Situ Apoptosis Kit (Green, FITC) (E-CK-A320) (Elabscience, Houston, TX, USA). Fluorescence images were obtained using a IX71 microscope (Olympus Corporation, Tokyo, Japan). Six views were randomly selected from two tumor sections per group. The cells were observed under the fluorescence microscope at 200× magnification. Positive cell counts were expressed as mean±standard error of the mean (SEM).

Statistical analysis. EZR (Saitama Medical Center, Jichi Medical University, Saitama, Japan), a graphical user interface for R (The R Foundation for Statistical Computing, Vienna, Austria), was used to perform all statistical analyses (22). The parametric test for between-group comparisons was the Tukey-Kramer HSD. Nonparametric tests were conducted using the Kruskal-Wallis test, and comparisons between groups were evaluated using the Steel-Dwass technique. Graphs show means±standard deviation (SD) or SEM. A p-value ≤0.05 was defined statistically significant.

Results

Treatment efficacy on DDLS-PDOX. The combination of CQ and RAPA arrested tumor growth in the DDLS PDOX compared to the untreated control (p=0.009) and was significantly more effective than RAPA alone (p=0.009). RAPA alone slowed tumor growth, but the difference was not statistically significant compared to the untreated control (p=0.2). CQ was not active alone (Figure 4). There was no significant difference in mouse body weight among the four groups (Figure 5).

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

Efficacy of drugs on the dedifferentiated liposarcoma patient-derived orthotopic xenograft model. Line graphs show tumor volume at the indicated times relative to that at the start of treatment. *p<0.05. Error bars: ±SEM. CQ, Chloroquine; RAPA, rapamycin.

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

Effect of drugs on relative mouse body weight. Bar graphs show body weight of mice from each group at day 15 relative to day 1 of treatment. Error bars: ±SD. CQ, Chloroquine; RAPA, rapamycin.

Histology of DDLS-PDOX. PDOX tissue in the untreated control and CQ alone consisted of dysplastic cells (Figure 6A and B). Treatment with RAPA alone as well as with the combination of CQ and RAPA showed fibrotic tissue with reduced cancer cell density (Figure 6C and D).

Figure 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 6.

Representative photomicrographs of H&E-stained dedifferentiated liposarcoma patient-derived orthotopic xenograft PDOX tissue sections. (A) Untreated control. (B) CQ-treated. (C) RAPA-treated. (D) Combination of CQ and RAPA. Magnification: 200×. Scale bar: 100 μm. CQ, Chloroquine; RAPA, rapamycin.

TUNEL assay of DDLS-PDOX. CQ combined with RAPA induced more apoptotic TUNEL-positive cells (24.33 cells) than the untreated control (3.33±1.60 cells); CQ alone (6.00±1.54 cells); and RAPA alone (7.00±1.93 cells) (p=0.02 for each comparison) (Figure 7 and Figure 8).

Figure 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 7.

TUNEL assay of apoptosis in the dedifferentiated liposarcoma patient-derived orthotopic xenograft tissue sections. 4’,6-Diamidino-2-phenylindole (DAPI) (blue) indicates cell nuclei and fluorescein isothiocyanate (FITC) (green) indicates apoptotic cells. Magnification: 200×. Scale bar: 25 μm. CQ, Chloroquine; RAPA, rapamycin.

Figure 8.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 8.

Apoptotic cell count by TUNEL assay. Cells were counted from 6 high-power microscopic fields from 2 TUNEL-stained tissue sections. *p<0.05. Error bars: ±SEM. CQ, Chloroquine; RAPA, rapamycin.

Discussion

Soft tissue sarcomas (STS) are rare mesenchymal malignant tumors that account for less than 1% of all adult malignancies. Liposarcoma is the most common histologic type of STS (23). In contrast to well-differentiated liposarcoma, which is less metastatic, the clinical behavior of DDLS is more aggressive, with a larger tendency for local recurrence and metastasis (2, 23). The five-year survival rate of well-differentiated is approximately 90%, whereas that of DDLS is 28% (1, 24). Because STS is a rare cancer with a small number of patients, the development of new drugs has lagged behind that for other, more frequent cancer types (25). Although pazopanib, eribulin, and trabectedin have been used to treat STS since 2010 (26-28), antitumor drugs such as doxorubicin and isophosphamide, developed in the 1970s and 1980s, are still the standard first-line treatment for STS (3-7).

Our previous studies have shown the efficacy of combining mTOR inhibitors with other antitumor agents against the PDOX mouse model of osteosarcoma (14, 15). In the present study, a PDOX mouse model of DDPL was established and was used to evaluate the anticancer efficacy of the mTOR inhibitor RAPA and the antimalarial drug CQ. The results demonstrated that the combination of CQ and RAPA was highly effective against the PDOX model of DDLP.

The PI3K/AKT/mTOR signaling system is activated in STS such as liposarcoma, Ewing’s sarcoma, rhabdomyosarcoma, and leiomyosarcoma (29-32). mTOR inhibitors are thought to have anticancer efficacy by reducing the activity of the signaling pathway and arresting the cell cycle in G1 phase (33). In the present study, however, RAPA alone had no significant tumor-suppression effect.

CQ can induce apoptosis of cancer cells by inhibiting autophagy (17). Autophagy is an important physiological mechanism that controls the breakdown of proteins and organelles and maintains homeostasis (34). In cancer cells, autophagy promotes their proliferation by recycling accumulated metabolites and positively regulates cancer cell metabolism (35). CQ appears to accumulate in lysosomes and may inhibit proteolysis of proteins that are imported into the cell by endocytosis and induce apoptosis in cancer cells (17, 35). For example, knockdown of LYSET, which is involved in targeting catabolic enzymes into lysosomes, greatly inhibits proteolysis in lysosomes, depriving the cancer cell of essential amino acids, including methionine, to which DDLS is addicted (36), and was shown to inhibit tumor growth (37).

In the present study, CQ alone did not significantly increase the number of apoptosis-positive cells and had no tumor suppressive effect. However, the combination of CQ and RAPA was found to significantly increase apoptosis-positive cells and enhance tumor suppression.

The efficacy of combining autophagy inhibitors with antineoplastic agents has been previously studied, and there are several reports that the combination can enhance apoptosis induction (18-20, 38-44). However, there were no reports that the combination of CQ and RAPA induces apoptosis and enhances tumor suppression in vivo, especially in a PDOX mouse model of DDLS. The present study showed for the first time that the combination of CQ and RAPA is effective against DDLS in vivo. This indicates CQ plus RAPA may be a potent clinical treatment for DDLS.

Conclusion

Combination therapy with CQ and RAPA, both FDA approved drugs, arrested a DDLS PDOX and demonstrated clinical potential for DDLS, a recalcitrant cancer. The present results suggest that treatment of DDLS patients with the combination of CQ and RAPA may be effective.

Acknowledgements

This paper is dedicated to the memory of A.R. Moossa, MD, Sun Lee, MD, Professor Li Jiaxi, Masaki Kitajima, MD, Shigeo Yagi, PhD and Jack Geller, MD. The present study was supported by the Robert M. Hoffman Foundation for Cancer Research.

Footnotes

  • Authors’ Contributions

    N.M. conceived the study, N.M., Y.A. Y. K. and K.O. performed the experiments and J.M. and R.M.H. provided scientific advice. N.M. wrote the paper and R.M.H. revised the paper.

  • Conflicts of Interest

    The Authors declare that they have no conflicts of interest in relation to this study. AntiCancer Inc. uses PDOX models for contract research.

  • Received August 4, 2022.
  • Revision received September 12, 2022.
  • Accepted September 28, 2022.
  • Copyright © 2022, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved

This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY-NC-ND) 4.0 international license (https://creativecommons.org/licenses/by-nc-nd/4.0).

References

  1. ↵
    1. Singer S,
    2. Antonescu CR,
    3. Riedel E and
    4. Brennan MF
    : Histologic subtype and margin of resection predict pattern of recurrence and survival for retroperitoneal liposarcoma. Ann Surg 238(3): 358-70; discussion 370-1, 2003. PMID: 14501502. DOI: 10.1097/01.sla.0000086542.11899.38
    OpenUrlCrossRefPubMed
  2. ↵
    1. Masaki N,
    2. Onozawa M,
    3. Inoue T,
    4. Kurobe M,
    5. Kawai K and
    6. Miyazaki J
    : Clinical features of multiply recurrent retroperitoneal liposarcoma: A single-center experience. Asian J Surg 44(1): 380-385, 2021. PMID: 33191070. DOI: 10.1016/j.asjsur.2020.10.015
    OpenUrlCrossRefPubMed
  3. ↵
    1. Elias A,
    2. Ryan L,
    3. Aisner J and
    4. Antman KH
    : Mesna, doxorubicin, ifosfamide, dacarbazine (MAID) regimen for adults with advanced sarcoma. Semin Oncol 17(2 Suppl 4): 41-49, 1990. PMID: 2110385.
    OpenUrlPubMed
    1. Santoro A,
    2. Tursz T,
    3. Mouridsen H,
    4. Verweij J,
    5. Steward W,
    6. Somers R,
    7. Buesa J,
    8. Casali P,
    9. Spooner D and
    10. Rankin E
    : Doxorubicin versus CYVADIC versus doxorubicin plus ifosfamide in first-line treatment of advanced soft tissue sarcomas: a randomized study of the European Organization for Research and Treatment of Cancer Soft Tissue and Bone Sarcoma Group. J Clin Oncol 13(7): 1537-1545, 1995. PMID: 7602342. DOI: 10.1200/JCO.1995.13.7.1537
    OpenUrlAbstract/FREE Full Text
    1. Judson I,
    2. Verweij J,
    3. Gelderblom H,
    4. Hartmann JT,
    5. Schöffski P,
    6. Blay JY,
    7. Kerst JM,
    8. Sufliarsky J,
    9. Whelan J,
    10. Hohenberger P,
    11. Krarup-Hansen A,
    12. Alcindor T,
    13. Marreaud S,
    14. Litière S,
    15. Hermans C,
    16. Fisher C,
    17. Hogendoorn PC,
    18. dei Tos AP,
    19. van der Graaf WT and European Organisation and Treatment of Cancer Soft Tissue and Bone Sarcoma Group
    : Doxorubicin alone versus intensified doxorubicin plus ifosfamide for first-line treatment of advanced or metastatic soft-tissue sarcoma: a randomised controlled phase 3 trial. Lancet Oncol 15(4): 415-423, 2014. PMID: 24618336. DOI: 10.1016/S1470-2045(14)70063-4
    OpenUrlCrossRefPubMed
    1. Yap BS,
    2. Baker LH,
    3. Sinkovics JG,
    4. Rivkin SE,
    5. Bottomley R,
    6. Thigpen T,
    7. Burgess MA,
    8. Benjamin RS and
    9. Bodey GP
    : Cyclophosphamide, vincristine, adriamycin, and DTIC (CYVADIC) combination chemotherapy for the treatment of advanced sarcomas. Cancer Treat Rep 64(1): 93-98, 1980. PMID: 7379060.
    OpenUrlPubMed
  4. ↵
    1. Schütte J,
    2. Mouridsen HT,
    3. Stewart W,
    4. Santoro A,
    5. van Oosterom AT,
    6. Somers R,
    7. Blackledge G,
    8. Verweij J,
    9. Dombernowsky P and
    10. Thomas D
    : Ifosfamide plus doxorubicin in previously untreated patients with advanced soft tissue sarcoma. The EORTC Soft Tissue and Bone Sarcoma Group. Eur J Cancer 26(5): 558-561, 1990. PMID: 2144740. DOI: 10.1016/0277-5379(90)90075-5
    OpenUrlCrossRefPubMed
  5. ↵
    1. Fu XY,
    2. Besterman JM,
    3. Monosov A and
    4. Hoffman RM
    : Models of human metastatic colon cancer in nude mice orthotopically constructed by using histologically intact patient specimens. Proc Natl Acad Sci U.S.A. 88(20): 9345-9349, 1991. PMID: 1924398. DOI: 10.1073/pnas.88.20.9345
    OpenUrlAbstract/FREE Full Text
  6. ↵
    1. Hoffman RM
    : Patient-derived orthotopic xenografts: better mimic of metastasis than subcutaneous xenografts. Nat Rev Cancer 15(8): 451-452, 2015. PMID: 26422835. DOI: 10.1038/nrc3972
    OpenUrlCrossRefPubMed
  7. ↵
    1. Furukawa T,
    2. Kubota T,
    3. Watanabe M,
    4. Kitajima M and
    5. Hoffman RM
    : Orthotopic transplantation of histologically intact clinical specimens of stomach cancer to nude mice: correlation of metastatic sites in mouse and individual patient donors. Int J Cancer 53(4): 608-612, 1993. PMID: 8436434. DOI: 10.1002/ijc.2910530414
    OpenUrlCrossRefPubMed
  8. ↵
    1. Saunders RN,
    2. Metcalfe MS and
    3. Nicholson ML
    : Rapamycin in transplantation: a review of the evidence. Kidney Int 59(1): 3-16, 2001. PMID: 11135052. DOI: 10.1046/j.1523-1755.2001.00460.x
    OpenUrlCrossRefPubMed
  9. ↵
    1. Motzer RJ,
    2. Escudier B,
    3. Oudard S,
    4. Hutson TE,
    5. Porta C,
    6. Bracarda S,
    7. Grünwald V,
    8. Thompson JA,
    9. Figlin RA,
    10. Hollaender N,
    11. Urbanowitz G,
    12. Berg WJ,
    13. Kay A,
    14. Lebwohl D,
    15. Ravaud A and RECORD-1 Study Group
    : Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet 372(9637): 449-456, 2008. PMID: 18653228. DOI: 10.1016/S0140-6736(08)61039-9
    OpenUrlCrossRefPubMed
  10. ↵
    1. Hay N and
    2. Sonenberg N
    : Upstream and downstream of mTOR. Genes Dev 18(16): 1926-1945, 2004. PMID: 15314020. DOI: 10.1101/gad.1212704
    OpenUrlAbstract/FREE Full Text
  11. ↵
    1. Oshiro H,
    2. Tome Y,
    3. Miyake K,
    4. Higuchi T,
    5. Sugisawa N,
    6. Kanaya F,
    7. Nishida K and
    8. Hoffman RM
    : Combination of CDK4/6 and mTOR inhibitors suppressed doxorubicin-resistant osteosarcoma in a patient-derived orthotopic xenograft mouse model: a translatable strategy for recalcitrant disease. Anticancer Res 41(7): 3287-3292, 2021. PMID: 34230123. DOI: 10.21873/anticanres.15115
    OpenUrlAbstract/FREE Full Text
  12. ↵
    1. Oshiro H,
    2. Tome Y,
    3. Miyake K,
    4. Higuchi T,
    5. Sugisawa N,
    6. Kanaya F,
    7. Nishida K and
    8. Hoffman RM
    : An mTOR and VEGFR inhibitor combination arrests a doxorubicin resistant lung metastatic osteosarcoma in a PDOX mouse model. Sci Rep 11(1): 8583, 2021. PMID: 33883561. DOI: 10.1038/s41598-021-87553-9
    OpenUrlCrossRefPubMed
  13. ↵
    1. Solomon VR and
    2. Lee H
    : Chloroquine and its analogs: a new promise of an old drug for effective and safe cancer therapies. Eur J Pharmacol 625(1-3): 220-233, 2009. PMID: 19836374. DOI: 10.1016/j.ejphar.2009.06.063
    OpenUrlCrossRefPubMed
  14. ↵
    1. Pascolo S
    : Time to use a dose of Chloroquine as an adjuvant to anti-cancer chemotherapies. Eur J Pharmacol 771: 139-144, 2016. PMID: 26687632. DOI: 10.1016/j.ejphar.2015.12.017
    OpenUrlCrossRefPubMed
  15. ↵
    1. Erkisa M,
    2. Aydinlik S,
    3. Cevatemre B,
    4. Aztopal N,
    5. Akar RO,
    6. Celikler S,
    7. Yilmaz VT,
    8. Ari F and
    9. Ulukaya E
    : A promising therapeutic combination for metastatic prostate cancer: Chloroquine as autophagy inhibitor and palladium(II) barbiturate complex. Biochimie 175: 159-172, 2020. PMID: 32497551. DOI: 10.1016/j.biochi.2020.05.010
    OpenUrlCrossRefPubMed
  16. ↵
    1. Guo XL,
    2. Li D,
    3. Hu F,
    4. Song JR,
    5. Zhang SS,
    6. Deng WJ,
    7. Sun K,
    8. Zhao QD,
    9. Xie XQ,
    10. Song YJ,
    11. Wu MC and
    12. Wei LX
    : Targeting autophagy potentiates chemotherapy-induced apoptosis and proliferation inhibition in hepatocarcinoma cells. Cancer Lett 320(2): 171-179, 2012. PMID: 22406827. DOI: 10.1016/j.canlet.2012.03.002
    OpenUrlCrossRefPubMed
  17. ↵
    1. Rangwala R,
    2. Chang YC,
    3. Hu J,
    4. Algazy KM,
    5. Evans TL,
    6. Fecher LA,
    7. Schuchter LM,
    8. Torigian DA,
    9. Panosian JT,
    10. Troxel AB,
    11. Tan KS,
    12. Heitjan DF,
    13. DeMichele AM,
    14. Vaughn DJ,
    15. Redlinger M,
    16. Alavi A,
    17. Kaiser J,
    18. Pontiggia L,
    19. Davis LE,
    20. O’Dwyer PJ and
    21. Amaravadi RK
    : Combined MTOR and autophagy inhibition: phase I trial of hydroxychloroquine and temsirolimus in patients with advanced solid tumors and melanoma. Autophagy 10(8): 1391-1402, 2014. PMID: 24991838. DOI: 10.4161/auto.29119
    OpenUrlCrossRefPubMed
  18. ↵
    1. Cardiff RD,
    2. Miller CH and
    3. Munn RJ
    : Manual hematoxylin and eosin staining of mouse tissue sections. Cold Spring Harb Protoc 2014(6): 655-658, 2014. PMID: 24890205. DOI: 10.1101/pdb.prot073411
    OpenUrlCrossRefPubMed
  19. ↵
    1. Kanda Y
    : Investigation of the freely available easy-to-use software ‘EZR’ for medical statistics. Bone Marrow Transplant 48(3): 452-458, 2013. PMID: 23208313. DOI: 10.1038/bmt.2012.244
    OpenUrlCrossRefPubMed
  20. ↵
    1. Gamboa AC,
    2. Gronchi A and
    3. Cardona K
    : Soft-tissue sarcoma in adults: An update on the current state of histiotype-specific management in an era of personalized medicine. CA Cancer J Clin 70(3): 200-229, 2020. PMID: 32275330. DOI: 10.3322/caac.21605
    OpenUrlCrossRefPubMed
  21. ↵
    1. Henricks WH,
    2. Chu YC,
    3. Goldblum JR and
    4. Weiss SW
    : Dedifferentiated liposarcoma: a clinicopathological analysis of 155 cases with a proposal for an expanded definition of dedifferentiation. Am J Surg Pathol 21(3): 271-281, 1997. PMID: 9060596. DOI: 10.1097/00000478-199703000-00002
    OpenUrlCrossRefPubMed
  22. ↵
    1. Yuan J,
    2. Li X and
    3. Yu S
    : Molecular targeted therapy for advanced or metastatic soft tissue sarcoma. Cancer Control 28: 10732748211038424, 2021. PMID: 34844463. DOI: 10.1177/10732748211038424
    OpenUrlCrossRefPubMed
  23. ↵
    1. van der Graaf WT,
    2. Blay JY,
    3. Chawla SP,
    4. Kim DW,
    5. Bui-Nguyen B,
    6. Casali PG,
    7. Schöffski P,
    8. Aglietta M,
    9. Staddon AP,
    10. Beppu Y,
    11. Le Cesne A,
    12. Gelderblom H,
    13. Judson IR,
    14. Araki N,
    15. Ouali M,
    16. Marreaud S,
    17. Hodge R,
    18. Dewji MR,
    19. Coens C,
    20. Demetri GD,
    21. Fletcher CD,
    22. Dei Tos AP,
    23. Hohenberger P, EORTC Soft Tissue and Bone Sarcoma Group and PALETTE study group
    : Pazopanib for metastatic soft-tissue sarcoma (PALETTE): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet 379(9829): 1879-1886, 2012. PMID: 22595799. DOI: 10.1016/S0140-6736(12)60651-5
    OpenUrlCrossRefPubMed
    1. Schöffski P,
    2. Chawla S,
    3. Maki RG,
    4. Italiano A,
    5. Gelderblom H,
    6. Choy E,
    7. Grignani G,
    8. Camargo V,
    9. Bauer S,
    10. Rha SY,
    11. Blay JY,
    12. Hohenberger P,
    13. D’Adamo D,
    14. Guo M,
    15. Chmielowski B,
    16. Le Cesne A,
    17. Demetri GD and
    18. Patel SR
    : Eribulin versus dacarbazine in previously treated patients with advanced liposarcoma or leiomyosarcoma: a randomised, open-label, multicentre, phase 3 trial. Lancet 387(10028): 1629-1637, 2016. PMID: 26874885. DOI: 10.1016/S0140-6736(15)01283-0
    OpenUrlCrossRefPubMed
  24. ↵
    1. Barone A,
    2. Chi DC,
    3. Theoret MR,
    4. Chen H,
    5. He K,
    6. Kufrin D,
    7. Helms WS,
    8. Subramaniam S,
    9. Zhao H,
    10. Patel A,
    11. Goldberg KB,
    12. Keegan P and
    13. Pazdur R
    : FDA approval summary: Trabectedin for unresectable or metastatic liposarcoma or leiomyosarcoma following an anthracycline-containing regimen. Clin Cancer Res 23(24): 7448-7453, 2017. PMID: 28774898. DOI: 10.1158/1078-0432.CCR-17-0898
    OpenUrlAbstract/FREE Full Text
  25. ↵
    1. Passacantilli I,
    2. Frisone P,
    3. De Paola E,
    4. Fidaleo M and
    5. Paronetto MP
    : hnRNPM guides an alternative splicing program in response to inhibition of the PI3K/AKT/mTOR pathway in Ewing sarcoma cells. Nucleic Acids Res 45(21): 12270-12284, 2017. PMID: 29036465. DOI: 10.1093/nar/gkx831
    OpenUrlCrossRefPubMed
    1. Laroche A,
    2. Chaire V,
    3. Algeo MP,
    4. Karanian M,
    5. Fourneaux B and
    6. Italiano A
    : MDM2 antagonists synergize with PI3K/mTOR inhibition in well-differentiated/dedifferentiated liposarcomas. Oncotarget 8(33): 53968-53977, 2017. PMID: 28903316. DOI: 10.18632/oncotarget.16345
    OpenUrlCrossRefPubMed
    1. Guenther MK,
    2. Graab U and
    3. Fulda S
    : Synthetic lethal interaction between PI3K/Akt/mTOR and Ras/MEK/ERK pathway inhibition in rhabdomyosarcoma. Cancer Lett 337(2): 200-209, 2013. PMID: 23684925. DOI: 10.1016/j.canlet.2013.05.010
    OpenUrlCrossRefPubMed
  26. ↵
    1. Hernando E,
    2. Charytonowicz E,
    3. Dudas ME,
    4. Menendez S,
    5. Matushansky I,
    6. Mills J,
    7. Socci ND,
    8. Behrendt N,
    9. Ma L,
    10. Maki RG,
    11. Pandolfi PP and
    12. Cordon-Cardo C
    : The AKT-mTOR pathway plays a critical role in the development of leiomyosarcomas. Nat Med 13(6): 748-753, 2007. PMID: 17496901. DOI: 10.1038/nm1560
    OpenUrlCrossRefPubMed
  27. ↵
    1. Gomez-Pinillos A and
    2. Ferrari AC
    : mTOR signaling pathway and mTOR inhibitors in cancer therapy. Hematol Oncol Clin North Am 26(3): 483-505, vii, 2012. PMID: 22520976. DOI: 10.1016/j.hoc.2012.02.014
    OpenUrlCrossRefPubMed
  28. ↵
    1. Klionsky DJ and
    2. Emr SD
    : Autophagy as a regulated pathway of cellular degradation. Science 290(5497): 1717-1721, 2000. PMID: 11099404. DOI: 10.1126/science.290.5497.1717
    OpenUrlAbstract/FREE Full Text
  29. ↵
    1. White E
    : Deconvoluting the context-dependent role for autophagy in cancer. Nat Rev Cancer 12(6): 401-410, 2012. PMID: 22534666. DOI: 10.1038/nrc3262
    OpenUrlCrossRefPubMed
  30. ↵
    1. Higuchi T,
    2. Han Q,
    3. Miyake K,
    4. Oshiro H,
    5. Sugisawa N,
    6. Tan Y,
    7. Yamamoto N,
    8. Hayashi K,
    9. Kimura H,
    10. Miwa S,
    11. Igarashi K,
    12. Bouvet M,
    13. Singh SR,
    14. Tsuchiya H and
    15. Hoffman RM
    : Combination of oral recombinant methioninase and decitabine arrests a chemotherapy-resistant undifferentiated soft-tissue sarcoma patient-derived orthotopic xenograft mouse model. Biochem Biophys Res Commun 523(1): 135-139, 2020. PMID: 31839218. DOI: 10.1016/j.bbrc.2019.12.024
    OpenUrlCrossRefPubMed
  31. ↵
    1. Pechincha C,
    2. Groessl S,
    3. Kalis R,
    4. de Almeida M,
    5. Zanotti A,
    6. Wittmann M,
    7. Schneider M,
    8. de Campos RP,
    9. Rieser S,
    10. Brandstetter M,
    11. Schleiffer A,
    12. Müller-Decker K,
    13. Helm D,
    14. Jabs S,
    15. Haselbach D,
    16. Lemberg MK,
    17. Zuber J and
    18. Palm W
    : Lysosomal enzyme trafficking factor LYSET enables nutritional usage of extracellular proteins. Science: eabn5637, 2022. PMID: 36074822. DOI: 10.1126/science.abn5637
    OpenUrlCrossRefPubMed
  32. ↵
    1. Ishibashi Y,
    2. Nakamura O,
    3. Yamagami Y,
    4. Nishimura H,
    5. Fukuoka N and
    6. Yamamoto T
    : Chloroquine enhances rapamycin-induced apoptosis in MG63 cells. Anticancer Res 39(2): 649-654, 2019. PMID: 30711941. DOI: 10.21873/anticanres.13159
    OpenUrlAbstract/FREE Full Text
    1. Shiratori H,
    2. Kawai K,
    3. Hata K,
    4. Tanaka T,
    5. Nishikawa T,
    6. Otani K,
    7. Sasaki K,
    8. Kaneko M,
    9. Murono K,
    10. Emoto S,
    11. Sonoda H and
    12. Nozawa H
    : The combination of temsirolimus and chloroquine increases radiosensitivity in colorectal cancer cells. Oncol Rep 42(1): 377-385, 2019. PMID: 31059051. DOI: 10.3892/or.2019.7134
    OpenUrlCrossRefPubMed
    1. Amaravadi RK,
    2. Yu D,
    3. Lum JJ,
    4. Bui T,
    5. Christophorou MA,
    6. Evan GI,
    7. Thomas-Tikhonenko A and
    8. Thompson CB
    : Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma. J Clin Invest 117(2): 326-336, 2007. PMID: 17235397. DOI: 10.1172/JCI28833
    OpenUrlCrossRefPubMed
    1. Bellodi C,
    2. Lidonnici MR,
    3. Hamilton A,
    4. Helgason GV,
    5. Soliera AR,
    6. Ronchetti M,
    7. Galavotti S,
    8. Young KW,
    9. Selmi T,
    10. Yacobi R,
    11. Van Etten RA,
    12. Donato N,
    13. Hunter A,
    14. Dinsdale D,
    15. Tirrò E,
    16. Vigneri P,
    17. Nicotera P,
    18. Dyer MJ,
    19. Holyoake T,
    20. Salomoni P and
    21. Calabretta B
    : Targeting autophagy potentiates tyrosine kinase inhibitor-induced cell death in Philadelphia chromosome-positive cells, including primary CML stem cells. J Clin Invest 119(5): 1109-1123, 2009. PMID: 19363292. DOI: 10.1172/JCI35660
    OpenUrlCrossRefPubMed
    1. Shimizu S,
    2. Takehara T,
    3. Hikita H,
    4. Kodama T,
    5. Tsunematsu H,
    6. Miyagi T,
    7. Hosui A,
    8. Ishida H,
    9. Tatsumi T,
    10. Kanto T,
    11. Hiramatsu N,
    12. Fujita N,
    13. Yoshimori T and
    14. Hayashi N
    : Inhibition of autophagy potentiates the antitumor effect of the multikinase inhibitor sorafenib in hepatocellular carcinoma. Int J Cancer 131(3): 548-557, 2012. PMID: 21858812. DOI: 10.1002/ijc.26374
    OpenUrlCrossRefPubMed
    1. Wang FT,
    2. Wang H,
    3. Wang QW,
    4. Pan MS,
    5. Li XP,
    6. Sun W and
    7. Fan YZ
    : Inhibition of autophagy by chloroquine enhances the antitumor activity of gemcitabine for gallbladder cancer. Cancer Chemother Pharmacol 86(2): 221-232, 2020. PMID: 32654071. DOI: 10.1007/s00280-020-04100-5
    OpenUrlCrossRefPubMed
  33. ↵
    1. Bryant KL,
    2. Stalnecker CA,
    3. Zeitouni D,
    4. Klomp JE,
    5. Peng S,
    6. Tikunov AP,
    7. Gunda V,
    8. Pierobon M,
    9. Waters AM,
    10. George SD,
    11. Tomar G,
    12. Papke B,
    13. Hobbs GA,
    14. Yan L,
    15. Hayes TK,
    16. Diehl JN,
    17. Goode GD,
    18. Chaika NV,
    19. Wang Y,
    20. Zhang GF,
    21. Witkiewicz AK,
    22. Knudsen ES,
    23. Petricoin EF 3rd.,
    24. Singh PK,
    25. Macdonald JM,
    26. Tran NL,
    27. Lyssiotis CA,
    28. Ying H,
    29. Kimmelman AC,
    30. Cox AD and
    31. Der CJ
    : Combination of ERK and autophagy inhibition as a treatment approach for pancreatic cancer. Nat Med 25(4): 628-640, 2019. PMID: 30833752. DOI: 10.1038/s41591-019-0368-8
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

In Vivo: 36 (6)
In Vivo
Vol. 36, Issue 6
November-December 2022
  • Table of Contents
  • Table of Contents (PDF)
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on In Vivo.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Chloroquine Combined With Rapamycin Arrests Tumor Growth in a Patient-derived Orthotopic Xenograft (PDOX) Mouse Model of Dedifferentiated Liposarcoma
(Your Name) has sent you a message from In Vivo
(Your Name) thought you would like to see the In Vivo web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
8 + 2 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Chloroquine Combined With Rapamycin Arrests Tumor Growth in a Patient-derived Orthotopic Xenograft (PDOX) Mouse Model of Dedifferentiated Liposarcoma
NORIYUKI MASAKI, YUSUKE AOKI, YUTARO KUBOTA, KOYA OBARA, JUN MIYAZAKI, ROBERT M. HOFFMAN
In Vivo Nov 2022, 36 (6) 2630-2637; DOI: 10.21873/invivo.12997

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Chloroquine Combined With Rapamycin Arrests Tumor Growth in a Patient-derived Orthotopic Xenograft (PDOX) Mouse Model of Dedifferentiated Liposarcoma
NORIYUKI MASAKI, YUSUKE AOKI, YUTARO KUBOTA, KOYA OBARA, JUN MIYAZAKI, ROBERT M. HOFFMAN
In Vivo Nov 2022, 36 (6) 2630-2637; DOI: 10.21873/invivo.12997
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Conclusion
    • Acknowledgements
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • Combination of Recombinant Methioninase With Rapamycin or Chloroquine Is Synergistic to Highly Inhibit Triple-negative Breast Cancer Cells In Vitro
  • The Combination of Tumor-targeting Salmonella typhimurium A1-R Plus the Autophagy-inhibitor Chloroquine Synergistically Eradicates HT1080 Fibrosarcoma Cells In Vitro and In Vivo
  • Targeting Autophagy With the Synergistic Combination of Chloroquine and Rapamycin as a Novel Effective Treatment for Well-differentiated Liposarcoma
  • Google Scholar

More in this TOC Section

  • Over-expression of Truncated IK Ameliorates Dinitrochlorobenzene-Induced Allergic Contact Dermatitis Lesions in BALB/c Mice
  • Association of Matrix Metalloproteinase-7 Genotypes With Nasopharyngeal Carcinoma Risk
  • Age- and Periodontal Disease Independent Correlation of Salivary Amino Acids
Show more Experimental Studies

Similar Articles

Keywords

  • Dedifferentiated liposarcoma
  • PDOX
  • patient-derived orthotopic xenograft
  • combination therapy
  • mTOR inhibitors
  • rapamycin
  • chloroquine
  • apoptosis
In Vivo

© 2025 In Vivo

Powered by HighWire