1,081
Views
0
CrossRef citations to date
0
Altmetric
Letter to the Editor

For HIPEC, synergistic effects of hyperthermia and doxorubicin are optimal when simultaneously combined

, , ORCID Icon, &
Pages 346-348 | Received 27 Feb 2020, Accepted 11 Mar 2020, Published online: 09 Apr 2020

Dear Sir,

We thank Dr. Chen and colleagues for their interest in our published findings, Increased uptake of doxorubicin by cells undergoing heat stress does not explain its synergistic cytotoxicity with hyperthermia [Citation1]. We have read their suggestions with interest. Below, we provide our response.

Clonogenic assay is the method of choice to quantify reproductive capacity in cells after exposure to ionizing radiation and other cytotoxic agents [Citation2]. It has become an established assay to measure biological effects of hyperthermia, alone or in conjunction with other modalities, by providing a method to quantify cellular response with thermal dose (exposure time at elevated temperature) [Citation2–7]. Given that it is an established tool to quantify the biological effects of hyperthermia and therapeutic enhancement with other modalities, it is the method of choice for the current study. Clonogenic assays integrate the culmination of multiple stress responses leading to loss of reproductive capacity in cells; however, they do not inform about specific pathway activation or mechanisms leading to reproductive cell death. Other assays measuring cell viability, metabolic function, membrane integrity, mechanisms of cell death, apoptosis, etc. are available and their use can provide insights into specific mechanisms; however, clonogenic assay is the only established method that provides a quantitative correlation with thermal dose and clinically relevant biological response. Thus, for quantitative comparisons of dose-response with hyperthermia and other modalities, the clonogenic assay remains the method of choice.

Hyperthermia and doxorubicin are pleiotropic cytotoxic agents affecting many cellular processes [Citation5–36]. As such, they induce multiple (dose dependent) cell stress mechanisms, with cell death resulting from a failure of repair/response to ensure recovery and continued proliferation. Our quantitative comparison establishes that combining hyperthermia with doxorubicin leads to synergistic therapeutic effects in the three colorectal cancer cell lines studied. We note that two of these cell lines are human and the third is murine. Thus, our results demonstrate that this synergistic effect is not necessarily limited to human or murine cells. Further, we emphasize that such quantitative comparisons are essential to substantiate claims of synergistic effects. Whether this synergistic relationship exists across all known colorectal cancer cell lines, or cell lines representing other cancers remains an open question. We suggest that future studies begin with a similarly quantitative assessment of the dose-response relationship between hyperthermia and doxorubicin as a beginning point for detailed inquiry into biological mechanisms.

The conclusion that the observed synergism is not explained by increased intracellular doxorubicin levels was ascertained with only one cell line (HCT116), demonstrating that other mechanisms are responsible for the effect in this cell line. Despite the obvious limitation, the conclusion remains valid for this cell line. In the manuscript, we make no claim as to its prevalence or general applicability to other cell lines. We clearly state that further study with multiple cell lines is warranted; however, depending on individual phenotype and genotype, a range of responses can be expected vis a vis pleiotropic agents. Nevertheless, it motivates further examination to elucidate how this relates to other cells. The results present opportunities for further exploration to develop a deeper understanding of the effects of doxorubicin and heat.

We agree that further study is needed to ascertain mechanistic details giving rise to this synergism. For such studies, a careful selection of the appropriate assay(s) to probe individual stress responses is necessary. We suggest the careful selection of additional cell lines, having documented deficiencies in stress response and repair mechanisms to complement inquiry into the mechanistic details. We hope that our findings will motivate a renewed interest in research probing specific cell stress response pathways leading to a deeper understanding of hyperthermia + doxorubicin across multiple cell lines.

Concerning timing between heat and doxorubicin treatments, we recognize there is a broader implication for hyperthermia and doxorubicin combinations on which the authors of the letter focus; and, we are aware and agree that complex cellular and physiologic processes continue after heating. In this work, we focused on short time intervals because these are relevant for HIPEC, a clinical procedure used to treat various peritoneal neoplasms with heated (to hyperthermic temperature) chemotherapeutics, one of these being doxorubicin [Citation37–43]. It was our intention to explore effects and time intervals relevant for this clinical procedure. Certainly, there is motivation to take a renewed interest in heat + doxorubicin combinations with varied timing that may be more relevant for other clinical applications. In their letter, Chen et al. mention several examples using modalities other than HIPEC. One of the mentioned applications is ablation of VX2 tumors. We note that ablation involves fundamentally different biological processes, and is a different clinical application [Citation6]. Ablation was not the focus of our study [Citation1]. It is, however, relevant for some modalities and we hope that results presented will prove informative for such future work.

In this work, we sought to identify whether there is a meaningful effect of sequence of treatment with heat + doxorubicin, and thermal dose in cellular response. In this context, our results provide information relevant for some clinical procedures, and we hope they will motivate others to reexamine the complex and potentially elegant interactions between these venerable agents.

Disclosure statement

No potential conflict of interest was reported by the author(s).

References

  • Sharma A, Özayral S, Caserto JS, et al. Increased uptake of doxorubicin by cells undergoing heat stress does not explain its synergistic cytotoxicity with hyperthermia. Int J Hyperthermia. 2019; 36:712–720.
  • Franken NA, Rodermond HM, Stap J, et al. Clonogenic assay of cells in vitro. Nat Protoc. 2006;1(5):2315–2319.
  • Sapareto SA, Dewey WC. Thermal dose determination in cancer therapy. Int J Radiat Oncol Biol Phys. 1984;10(6):787–800.
  • Gillette EL. Clinical use of thermal enhancement and therapeutic gain for hyperthermia combined with radiation or drugs. Cancer Res. 1984;44:4836s–4841s.
  • Sapareto SA, Hopwood LE, Dewey WC, et al. Effects of hyperthermia on survival and progression of Chinese hamster ovary cells. Cancer Res. 1978;38:393–400.
  • Dewhirst MW, Viglianti BL, Lora-Michiels M, et al. Basic principles of thermal dosimetry and thermal thresholds for tissue damage from hyperthermia. Int J Hyperthermia. 2003;19(3):267–294.
  • Horsman MR, Overgaard J. Hyperthermia: a potent enhancer of radiotherapy. Clin Oncol. 2007;19(6):418–426.
  • Issels R, Kampmann E, Kanaar R, et al. Hallmarks of hyperthermia in driving the future of clinical hyperthermia as targeted therapy: translation into clinical application. Int. J. Hyperthermia. 2016;32(1):89–95.
  • Oei AL, Vriend LEM, Crezee J, et al. Effects of hyperthermia on DNA repair pathways: one treatment to inhibit them all. Radiat Oncol. 2015;10(1):13.
  • Roti Roti JL. Heat-induced alterations of nuclear protein associations and their effects on DNA repair and replication. Int J Hyperthermia. 2007;23(1):3–15.
  • Elming PB, Sørensen BS, Oei AL, Franken NAP, et al. Hyperthermia: the optimal treatment to overcome radiation resistant hypoxia. Cancers. 2019;11(1):60–20.
  • Krawczyk PM, Eppink B, Essers J, et al. Mild hyperthermia inhibits homologous recombination, induces BRCA2 degradation, and sensitizes cancer cells to poly (ADP-ribose) polymerase-1 inhibition. Proc Natl Acad Sci. 2011;108(24):9851–9856.
  • Mace TA, Zhong L, Kokolus KM, et al. Effector CD8+ T cell IFN-γ production and cytotoxicity are enhance by mild hyperthermia. Int J Hyperthermia. 2012;28(1):9–18.
  • Toraya-Brown S, Fiering S. Local tumour hyperthermia as immunotherapy for metastatic cancer. Int J Hyperthermia. 2014;30(8):531–539.
  • Repasky EA, Evans SS, Dewhirst MW. Temperature matters! And why it should matter to tumor immunologists. Cancer Immunol. Res. 2013;1(4):210–216.
  • Frey B, Weiss E-M, Rubner Y, et al. Old and new facts about hyperthermia-induced modulations of the immune system. Int J Hyperthermia. 2012;28(6):528–542.
  • Knippertz I, Stein MF, Dorrie J, et al. Mild hyperthermia enhances human monocyte-derived dendritic cell functions and offers potential for applications in vaccination strategies. Int J Hyperthermia. 2011;27(6):591–603.
  • Hatzfeld-Charbonnier AS, Lasek A, Castera L, et al. Influence of heat stress on human monocyte-derived dendritic cell functions with immunotherapeutic potential for antitumor vaccines. J Leukocyte Biol. 2007;81(5):1179–1187.
  • Zhang Y, Zhang W, Geng C, et al. Thermal ablation versus conventional regional hyperthermia has greater anti-tumor activity against melanoma in mice by upregulating CD4+ cells and enhancing IL-2 secretion. Prog Nat Sci. 2009;19:1699–1704.
  • Moon EJ, Sonveaux P, Porporato PE, et al. NADPH oxidase-mediated reactive oxygen species production activates hypoxia-inducible factor-1 (HIF-1) via the ERK pathway after hyperthermia treatment. Proc Natl Acad Sci USA. 2010;107(47):20477–20482.
  • Hildebrandt B, Wust P, Ahlers O, et al. The cellular and molecular basis of hyperthermia. Crit Rev Oncol Hematol. 2002;43(1):33–56.
  • van den Tempel N, Horsman MR, Kanaar R. Improving efficacy of hyperthermia in oncology by exploiting biological mechanisms. Int J Hyperthermia. 2016;32(4):446–454.
  • Kim S, Kim JH. Lethal effect of adriamycin on the division cycle of HeLa cells. Cancer Res. 1972;32(2):323–325.
  • Meriwether WD, Bachur NR. Inhibition of DNA and RNA metabolism by daunorubicin and adriamycin in L1210 mouse leukemia. Cancer Res. 1972;32(6):1137–1142.
  • Wang JJ, Chervinsky DS, Rosen JM. Comparative biochemical studies of adriamycin and daunomycin in leukemic cells. Cancer Res. 1972;32(3):511–515.
  • Momparler RL, Karon M, Siegel SE, et al. Effect of adriamycin on DNA, RNA, and protein synthesis in cell-free systems and intact cells. Cancer Res. 1976;36(8):2891–2895.
  • Bachur NR, Gordon SL, Gee MV. Anthracycline antibiotic augmentation of microsomal electron transport and free radical formation. Mol Pharmacol. 1977;13(5):901–910.
  • Berlin V, Haseltine WA. Reduction of adriamycin to a semiquinone-free radical by NADPH cytochrome P-450 reductase produces DNA cleavage in a reaction mediated by molecular oxygen. J Biol Chem. 1981;256(10):4747–4756.
  • Bates DA, Winterbourn CC. Deoxyribose breakdown by the adriamycin semiquinone and H2O2: evidence for hydroxyl radical participation. FEBS Lett. 1982;145(1):137–142.
  • Griffin-Green EA, Zaleska MM, Erecińska M. Adriamycin-induced lipid peroxidation in mitochondria and microsomes. Biochem Pharmacol. 1988;37(16):3071–3077.
  • Sinha BK, Trush MA, Kennedy KA, et al. Enzymatic activation and binding of adriamycin to nuclear DNA. Cancer Res. 1984;44(7):2892–2896.
  • Skladanowski A, Konopa J. Relevance of interstrand DNA crosslinking induced by anthracyclines for their biological activity. Biochem Pharmacol. 1994;47(12):2279–2287.
  • Tewey KM, Rowe TC, Yang L, et al. Adriamycin-induced DNA damage mediated by mammalian DNA topoisomerase II. Science. 1984;226(4673):466–468.
  • Capranico G, Dasdia T, Zunino F. Comparison of doxorubicin‐induced DNA damage in doxorubicin‐sensitive and‐resistant P388 murine leukemia cells. Int J Cancer. 1986;37(2):227–231.
  • Lawrence TS. Reduction of doxorubicin cytotoxicity by ouabain: correlation with topoisomerase-induced DNA strand breakage in human and hamster cells. Cancer Res. 1988;48(3):725–730.
  • Pang B, Qiao X, Janssen L, et al. Drug-induced histone eviction from open chromatin contributes to the chemotherapeutic effects of doxorubicin. Nat Commun. 2013;4(1):1908.
  • Cotte E, Glehen O, Mohamed F, et al. Cytoreductive surgery and intraperitoneal chemohyperthermia for chemoresistant and recurrent advanced epithelial ovarian cancer: prospective study of 81 patients. World J Surg. 2007;31(9):1813–1820.
  • Harrison LE, Bryan M, Pliner L, et al. Phase I trial of pegylated liposomal doxorubicin with hyperthermic intraperitoneal chemotherapy in patients undergoing cytoreduction for advanced intra-abdominal malignancy. Ann Surg Oncol. 2008;15(5):1407–1413.
  • Hompes D, D’Hoore A, Cutsem E, et al. The treatment of peritoneal carcinomatosis of colorectal cancer with complete cytoreductive surgery and hyperthermic intraperitoneal peroperative chemotherapy (HIPEC) with oxaliplatin: a Belgian multicentre prospective phase II clinical study. Ann Surg Oncol. 2012;19(7):2186–2194.
  • Polom K, Marano L, Roviello G, et al. Evolution and emerging future of cytoreducxtive surgery and hyperthermic intraperitoneal chemoperfusion in gastric cancer: from treating the incurable to preventing recurrence. Int J Hyperther. 2016;32(2):173–179.
  • González-Moreno S, González-Bayón LA, Ortega-Pérez G. Hyperthermic intraperitoneal chemotherapy: rationale and technique. WJGO. 2010;2(2):68.
  • Wang CC, Chen F, Kim E, et al. Thermal sensitization through ROS modulation: a strategy to improve the efficacy of hyperthermic intraperitoneal chemotherapy. Surgery. 2007;142(3):384–392.
  • Lehmann K, Rickenbacher A, Jang JH, et al. New insight into hyperthermic intraperitoneal chemotherapy: induction of oxidative stress dramatically enhanced tumor killing in in vitro and in vivo models. Ann Surg. 2012;256(5):730–738.