OBM Transplantation (ISSN 2577-5820) is an international peer-reviewed Open Access journal published quarterly online by LIDSEN Publishing Inc., which covers all evidence-based scientific studies related to transplantation, including: transplantation procedures and the maintenance of transplanted tissues or organs; assimilation of grafted tissue and the reconstitution of removed organs or parts of organs; transplantation of heart, lung, kidney, liver, pancreatic islets and bone marrow, etc. Areas related to clinical and experimental transplantation are also of interest.

OBM Transplantation is committed to rapid review and publication, and we aim at serving the international transplant community with high accessibility as well as relevant and high quality content.

We welcome original clinical studies as well as basic science, reviews, short reports/rapid communications, case reports, opinions, technical notes, book reviews as well as letters to the editor. 

Indexing:

Publication Speed (median values for papers published in 2023): Submission to First Decision: 6.7 weeks; Submission to Acceptance: 14.4 weeks; Acceptance to Publication: 6 days (1-2 days of FREE language polishing included)

Current Issue: 2024  Archive: 2023 2022 2021 2020 2019 2018 2017
Open Access Editorial

The Role of Cytochrome P450 2J2 in Cancer: Cell Protector, Therapeutic Target, or Prognostic Marker?

Ibrahim El-Serafi 1,2,3,*, Sandra Oerther 2,4, Ying Zhao 2,4, Moustapha Hassan 2,4

  1. Basic Medical Sciences Department, College of Medicine, Ajman University, Ajman, UAE

  2. Experimental Cancer Medicine (ECM), Clinical Research Centre, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden

  3. Department of Biochemistry, Faculty of Medicine, Port-Said University, Port-Said, Egypt

  4. Clinical research center (KFC), Novum, Karolinska University Hospital-Huddinge, Stockholm, Sweden

Correspondence: Dr. Ibrahim El-Serafi

Academic Editor: Haval Shirwan

Special Issue: Allogeneic Stem Cell Transplantation

Received: July 08, 2022 | Accepted: July 20, 2022 | Published: July 27, 2022

OBM Transplantation 2022, Volume 6, Issue 3, doi:10.21926/obm.transplant.2203163

Recommended citation: El-Serafi I, Oerther S, Zhao Y, Hassan M. The Role of Cytochrome P450 2J2 in Cancer: Cell Protector, Therapeutic Target, or Prognostic Marker?. OBM Transplantation 2022; 6(3): 163; doi:10.21926/obm.transplant.2203163.

© 2022 by the authors. This is an open access article distributed under the conditions of the Creative Commons by Attribution License, which permits unrestricted use, distribution, and reproduction in any medium or format, provided the original work is correctly cited.

Abstract

Cytochrome P450 2J2 (CYP2J2) is one of the recently discovered enzymes that is involved in the metabolism of several drugs. It is mainly an extrahepatic enzyme which can, to some extent, explain some the drugs’ side effects such as cyclophosphamide (Cy). Cyclophosphamide is an alkylating agent that is commonly used in conditioning prior to stem cell transplantation. It is a prodrug that is bio-activated by several CYPs including CYP2J2. Cy is known to have several side effects and its extrahepatic activation by CYP2J2 may explain the mechanism of some of these side effects. On the other hand, CYP2J2 is overexpressed in several types of cancer and its inhibition (e.g. by Cy) reduces the cancer cells survival. Taken together, CYP2J2 is considered as a potential therapeutic target and prognostic marker for cancer patients in addition to its role in drugs extrahepatic metabolism.

Graphical abstract

Click to view original image

Keywords

Stem cell transplantation; cyclophosphamide; cytochrome P450; CYP2J2; conditioning

Recently, the 60th anniversary of the first successful stem cell transplantation, performed by the Nobel Prize laureate E. Donnall Thomas in 1957 [1], was celebrated. In this editorial letter, we discussed the role and importance of cytochrome P450 2J2 (CYP2J2) in cyclophosphamide (Cy) bio-activation to highlight and explain several drug interactions in addition to the acute and chronic treatment toxicity reported after treatment with Cy.

Briefly, hematopoietic stem cell transplantation (HSCT) is a curative treatment for malignant diseases, such as leukemia, lymphomas, myeloma, and some solid tumors, and non-malignant diseases, such as metabolic and genetic disorders, thalassemia, sickle cell anemia, and aplastic anemia [2].

Hematopoietic stem cell transplantation is performed in several steps starting with a conditioning regimen, which eliminates malignant cells, provides a space for the donor cells, and provides enough immune suppression of the host immune system to avoid graft rejection. The conditioning protocol before conducting HSCT is based on administering chemotherapy (high doses of cytostatic drugs) along with or without total body irradiation (TBI). Immediately after conditioning, the patient receives either syngeneic or allogeneic stem cells. This causes the patient to experience an aplastic/neutropenic phase followed by a post-engraftment period.

Total body irradiation with or without cytostatic drugs was used for many years and is associated with acute and chronic complications[3,4], such as stomatitis, enteritis, and infection-related complications. Furthermore, the development of secondary leukemia post‑transplantation in donor-derived cells has been reported[5]. TBI is also associated with late complications such as secondary malignancies, including solid tumors, cataracts, CNS damage in pediatric patients, impaired growth, and endocrine dysfunction [6,7]. To avoid TBI, especially in pediatric patients, chemotherapy-based conditioning regimen protocols have been developed [6]. Nitrogen mustard was developed during World War II and was the first alkylating agent to be used for treating lymphoma. Inter-crosslinking and intra-crosslinking of DNA are the underlying mechanisms by which nitrogen mustard leads to cell death of rapidly dividing cells, including cancer cells. Ideally, TBI and cytostatic drugs should exert minimal toxic effects on normal host cells and tissues [8].

Cyclophosphamide (Cy) is an alkylating agent that was approved for medical use in 1959 and still plays a key role in cancer therapy. Cyclophosphamide is commonly used at high doses as a part of the conditioning regimen before conducting HSCT [6]. Besides its role in conditioning, Cy is used widely in moderate doses to treat malignancies, such as ovarian cancer, breast cancer, small-cell lung cancer, and neuroblastoma. Moreover, Cy at low doses is also used as an immunosuppressive drug for treating rheumatoid arthritis, systemic lupus erythematosus, Sjögren´s syndrome, and other autoimmune diseases. Recently, Cy was used for treatment after HSCT to prevent rejection and decrease the severity of graft-versus-host disease (GVHD) [7]. As an alkylating agent, Cy covalently binds to the guanine-N-7 base of DNA. This interaction enhances DNA damage that triggers apoptosis when the cellular machinery cannot be repaired.

Cyclophosphamide is a prodrug that needs to be bio-activated to exert its effect. It is metabolized via several CYPs into different metabolites but mainly into the active metabolite 4‑hydroxy‑cyclophosphamide (4‑OH-Cy), corresponding to more than 80% of the total dose of Cy [9]. Most of the final products are metabolized into substances that have cytotoxic effects (Figure 1) [10,11,12].

Click to view original image

Figure 1 The metabolic pathway of cyclophosphamide.

Cyclophosphamide is metabolized mainly through cytochrome CYP2B6 [10,13], while other enzymes, such as CYP3A4, CYP3A5, CYP2C9, and CYP2C19, are involved but to a lesser extent [114,15].

Interestingly, CYP2J2 is involved in the metabolism of Cy [16]. It is expressed mainly in the extrahepatic compartments, i.e., in the intestine and the cardiovascular system [17,18,19]. High CYP2J2 activity in the intestine contributes to the first-pass metabolism of several drugs [20,21,22], while in the human heart, CYP2J2 is responsible for the epoxidation of endogenous arachidonic acid to four regioisomeric epoxyeicosatrienoic acids (EETs) that are released in response to specific stimuli, such as ischemia [23].

Furthermore, CYP2J2 is related to malignancy. Several studies have reported that CYP2J2 is highly expressed in human-derived and mouse-derived malignant hematological cell lines, ovarian and lung cancer cell lines, as well as peripheral blood and bone marrow cells of leukemia patients [24]. High expression of CYP2J2 is associated with accelerated proliferation and attenuated apoptosis in different malignancies, including metastases [24,25,26]. Additionally, the inhibition of CYP2J2 is associated with suppressed proliferation of human cancer cells both in vitro and in vivo [27], which implies that CYP2J2 has a protective mechanism for cancer cell survival, and its inhibition was shown to suppress the proliferation of cancer cells [27].

A study found that Cy-treatment upregulated the expression of CYP2J2; however, the inter-individual variation was high [28]. Despite the high variability, the expression of CYP2J2 was significantly correlated with the bio-activation of Cy, as determined by the concentration ratio of 4-OH-Cy/Cy [28]. The inhibition of CYP2J2 in HL-60 cells can decrease the formation of 4‑OH‑Cy and concomitantly increase cell viability, which supports the role of CYP2J2 in Cy bioactivation [24]. In some populations, the ratio Vmax/Km (Vmax is the maximum velocity of the reaction where all the enzymes are saturated with the substrate and Km is the concentration of substrate at which half of the maximum velocity is achieved) for CYP2J2 was found to be higher than that obtained for CYP2B6 [29]. This suggested that CYP2J2 might be the predominant enzyme responsible for Cy bioactivation in these patients and not CYP2B6, as previously reported [10,13].

The above-mentioned studies, along with our observations, highlight the importance of the role of CYP2J2 in Cy bio-activation and its implication on drug-related toxicities, especially specific organ toxicities where CYP2J2 is highly expressed, for example, in the urinary bladder, heart, and intestine. Hemorrhagic cystitis or impairment in the removal of water from the urinary bladder and changes in the permeability of the intestine accompanied by diarrhea as a marker for intestinal barrier dysfunction are some examples of toxicity that occur after Cy-treatment in several patients [30]. A high dose of Cy is associated with acute cardiac toxicity, such as heart failure or a decrease in systolic function [31]. Damage to the endothelial cells in the mesenteric artery is also associated with Cy-treatment [32,33].

These toxic effects, along with the alloreactivity following HSCT, suggest that the bio-activation of Cy to 4-OH-Cy via CYP2J2 might also contribute to the occurrence of GVHD observed in transplanted patients. However, further studies are required to investigate the effect of the inhibition of CYP2J2 on the bio-activation of Cy to determine drug efficacy and clinical outcomes. Moreover, drugs metabolized via CYP2J2 and concomitantly used during Cy-conditioning might significantly alter Cy kinetics, along with the exposure, treatment efficacy, and toxicity of Cy, which might explain some of the major side effects reported after HSCT.

To summarize, the CYP2J2 gene is highly expressed in several malignancies. Further studies need to be conducted urgently to elucidate the role of CYP2J2 as a biomarker for different malignancies, treatment efficacy, personalized medicine, and/or as a target for cancer therapy.

Acknowledgments

The authors express their gratitude to Ajman University UAE, the Swedish children Cancer society (BCF) and Cimed for their support.

Author Contributions

IES: Conceptualization, formal analysis, visualization, writing–original draft, and editing. SO: Conceptualization, formal analysis, writing–original draft, and editing. YZ: investigation and editing. MH: Conceptualization, formal analysis, investigation, writing–original draft, and editing.

Competing Interests

The authors have declared that no competing interests exist.

References

  1. Thomas ED, Lochte Jr HL, Lu WC, Ferrebee JW. Intravenous infusion of bone marrow in patients receiving radiation and chemotherapy. N Engl J Med. 1957; 257: 491-496. [CrossRef]
  2. Riley RS, Idowu M, Chesney A, Zhao S, McCarty J, Lamb LS, et al. Hematologic aspects of myeloablative therapy and bone marrow transplantation. J Clin Lab Anal. 2005; 19: 47-79. [CrossRef]
  3. Achari R, Das A, Mahata A. Total body irradiation in stem cell transplant. Contemp Bone Marrow Transplant. 2020:1-8.
  4. Sabloff M, Tisseverasinghe S, Babadagli ME, Samant R. Total body irradiation for hematopoietic stem cell transplantation: What can we agree on? Curr Oncol. 2021; 28: 903-917.
  5. Baker KS, Leisenring WM, Goodman PJ, Ermoian RP, Flowers ME, Schoch G, et al. Total body irradiation dose and risk of subsequent neoplasms following allogeneic hematopoietic cell transplantation. Blood. 2019; 133: 2790-2799. [CrossRef]
  6. Kondo N, Takahashi A, Ono K, Ohnishi T. DNA damage induced by alkylating agents and repair pathways. J Nucleic Acids. 2010; 2010: 543531. [CrossRef]
  7. Luznik L, Fuchs EJ. High-dose, post-transplantation cyclophosphamide to promote graft-host tolerance after allogeneic hematopoietic stem cell transplantation. Immunol Res. 2010; 47: 65-77. [CrossRef]
  8. Burt RK, Loh Y, Pearce W, Beohar N, Barr WG, Craig R, et al. Clinical applications of blood-derived and marrow-derived stem cells for nonmalignant diseases. JAMA. 2008; 299: 925-936. [CrossRef]
  9. Sladek NE. Metabolism of oxazaphosphorines. Pharmacol Ther. 1988; 37: 301-355. [CrossRef]
  10. Cho JY, Lim HS, Chung JY, Yu KS, Kim JR, Shin SG, et al. Haplotype structure and allele frequencies of CYP2B6 in a Korean population. Drug Metab Dispos. 2004; 32: 1341-1344. [CrossRef]
  11. Chang TK, Weber GF, Crespi CL, Waxman DJ. Differential activation of cyclophosphamide and ifosphamide by cytochromes P-450 2B and 3A in human liver microsomes. Cancer Res. 1993; 53: 5629-5637.
  12. Ren S, Yang JS, Kalhorn TF, Slattery JT. Oxidation of cyclophosphamide to 4-hydroxycyclophosphamide and deschloroethylcyclophosphamide in human liver microsomes. Cancer Res. 1997; 57: 4229-4235.
  13. Raccor BS, Claessens AJ, Dinh JC, Park JR, Hawkins DS, Thomas SS, et al. Potential contribution of cytochrome P450 2B6 to hepatic 4-hydroxycyclophosphamide formation in vitro and in vivo. Drug Metab Dispos. 2012; 40: 54-63. [CrossRef]
  14. Afsar NA, Ufer M, Haenisch S, Remmler C, Mateen A, Usman A, et al. Relationship of drug metabolizing enzyme genotype to plasma levels as well as myelotoxicity of cyclophosphamide in breast cancer patients. Eur J Clin Pharmacol. 2012; 68: 389-395. [CrossRef]
  15. Fernandes BJ, Miranda Silva CD, Andrade JM, Matthes ÂD, Coelho EB, Lanchote VL. Pharmacokinetics of cyclophosphamide enantiomers in patients with breast cancer. Cancer Chemother Pharmacol. 2011; 68: 897-904. [CrossRef]
  16. Ma J, Ramachandran S, Fiedorek Jr FT, Zeldin DC. Mapping of the CYP2J cytochrome P450 genes to human chromosome 1 and mouse chromosome 4. Genomics. 1998; 49: 152-155. [CrossRef]
  17. DeLozier TC, Kissling GE, Coulter SJ, Dai D, Foley JF, Bradbury JA, et al. Detection of human CYP2C8, CYP2C9, and CYP2J2 in cardiovascular tissues. Drug Metab Dispos. 2007; 35: 682-688. [CrossRef]
  18. Node K, Huo Y, Ruan X, Yang B, Spiecker M, Ley K, et al. Anti-inflammatory properties of cytochrome P450 epoxygenase-derived eicosanoids. Science. 1999; 285: 1276-1279. [CrossRef]
  19. Xu X, Zhang XA, Wang DW. The roles of CYP450 epoxygenases and metabolites, epoxyeicosatrienoic acids, in cardiovascular and malignant diseases. Adv Drug Deliv Rev. 2011; 63: 597-609. [CrossRef]
  20. Hashizume T, Imaoka S, Mise M, Terauchi Y, Fujii T, Miyazaki H, et al. Involvement of CYP2J2 and CYP4F12 in the metabolism of ebastine in human intestinal microsomes. J Pharmacol Exp Ther. 2002; 300: 298-304. [CrossRef]
  21. Lee CA, Neul D, Clouser-Roche A, Dalvie D, Wester MR, Jiang Y, et al. Identification of novel substrates for human cytochrome P450 2J2. Drug Metab Dispos. 2010; 38: 347-356. [CrossRef]
  22. Matsumoto S, Hirama T, Matsubara T, Nagata K, Yamazoe Y. Involvement of CYP2J2 on the intestinal first-pass metabolism of antihistamine drug, astemizole. Drug Metab Dispos. 2002; 30: 1240-1245. [CrossRef]
  23. Noshita N, Sugawara T, Hayashi T, Lewén A, Omar G, Chan PH. Copper/zinc superoxide dismutase attenuates neuronal cell death by preventing extracellular signal-regulated kinase activation after transient focal cerebral ischemia in mice. J Neurosci. 2002; 22: 7923-7930. [CrossRef]
  24. Chen C, Wei X, Rao X, Wu J, Yang S, Chen F, et al. Cytochrome P450 2J2 is highly expressed in hematologic malignant diseases and promotes tumor cell growth. J Pharmacol Exp Ther. 2011; 336: 344-355. [CrossRef]
  25. Freedman RS, Wang E, Voiculescu S, Patenia R, Bassett Jr RL, Deavers M, et al. Comparative analysis of peritoneum and tumor eicosanoids and pathways in advanced ovarian cancer. Clin Cancer Res. 2007; 13: 5736-5744. [CrossRef]
  26. Jiang JG, Ning YG, Chen C, Ma D, Liu ZJ, Yang S, et al. Cytochrome P 450 epoxygenase promotes human cancer metastasis. Cancer Res. 2007; 67: 6665-6674. [CrossRef]
  27. Chen C, Li G, Liao W, Wu J, Liu L, Ma D, et al. Selective inhibitors of CYP2J2 related to terfenadine exhibit strong activity against human cancers in vitro and in vivo. J Pharmacol Exp Ther. 2009; 329: 908-918. [CrossRef]
  28. El-Serafi I, Fares M, Abedi-Valugerdi M, Afsharian P, Moshfegh A, Terelius Y, et al. Cytochrome P450 2J2, a new key enzyme in cyclophosphamide bioactivation and a potential biomarker for hematological malignancies. Pharmacogenomics J. 2015; 15: 405-413. [CrossRef]
  29. Nguyen TA, Tychopoulos M, Bichat F, Zimmermann C, Flinois JP, Diry M, et al. Improvement of cyclophosphamide activation by CYP2B6 mutants: From in silico to ex vivo. Mol Pharmacol. 2008; 73: 1122-1133. [CrossRef]
  30. Russo F, Linsalata M, Clemente C, D’Attoma B, Orlando A, Campanella G, et al. The effects of fluorouracil, epirubicin, and cyclophosphamide (FEC60) on the intestinal barrier function and gut peptides in breast cancer patients: An observational study. BMC Cancer. 2013; 13: 56. [CrossRef]
  31. Gharib MI, Burnett AK. Chemotherapy-induced cardiotoxicity: Current practice and prospects of prophylaxis. Eur J Heart Fail. 2002; 4: 235-242. [CrossRef]
  32. Al-Hashmi S, Boels PJ, Zadjali F, Sadeghi B, Sällström J, Hultenby K, et al. Busulphan-cyclophosphamide cause endothelial injury, remodeling of resistance arteries and enhanced expression of endothelial nitric oxide synthase. PLoS One. 2012; 7: e30897.
  33. Chow EJ, Wong K, Lee SJ, Cushing-Haugen KL, Flowers ME, Friedman DL, et al. Late cardiovascular complications after hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2014; 20: 794-800. [CrossRef]
Newsletter
Download PDF Download Citation
0 0

TOP