#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Methotrexate update 2014: 70 years in autoimmunity and cancer treatment


Authors: M. Řiháček 1,2;  I. Řiháček 1,3;  L. Zdražilová-Dubská 1,2,4;  K. Pilátová 1,2
Authors‘ workplace: Faculty of Medicine, Masaryk University, Kamenice, Brno, Czech RepublicHead prof. MUDr. J. Mayer, CSc. 1;  Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech RepublicHead doc. MUDr. D. Valík, Ph. D. 2;  Department of Pharmacology – ACIU, Faculty of Medicine, Masaryk University, Kamenice, Brno, Czech RepublicHead MUDr. R. Demlová, Ph. D. 4;  nd Department of Internal Medicine, St. Anne’s University Hospital, Brno. Czech RepublicHead prof. MUDr. M. Souček, CSc. 32
Published in: Čes-slov Pediat 2014; 69 (3): 161-167.
Category: Review

Overview

Immunosupressive and antineoplastic activity brought methotrexate to its clinical use many years ago. Although modern antitumour and antiinflammatory drugs and new antifolates have challenged methotrexate, its use in rheumatoid arthritis and in some types of pediatric and adult malignancies remains a gold standard. The principal main mechanism of action of methotrexate is competitive inhibition of dihydrofolate reductase. Recently, other pharmacodynamic effects have been described. Potent inflammation inducers such as leukotriene-B4 and polyamine synthesis were found to be reduced by methotrexate treatment. Jun N-terminal kinase and B cell lymphoma 2 gene are downregulated by methotrexate. Its metabolite, 7-hydroxymethotrexate and also its intracellular polyglutamated forms show a different spectrum of inhibitory activity as well as potency towards dihydrofolate reductase and other enzymes. In this review, we summarize recent information data on methotrexate in both fields of its use, oncology and rheumatology. We also discuss new biomarkers of toxicity for further improvement of therapeutic protocols.

Keywords:
methotrexate, autoimmunity, neoplasms, biological markers, toxicity, adverse effects

Introduction

Methotrexate (MTX) is an antineoplastic agent first reported by Sidney Farber in 1948 in children with leukaemia [1]. Antiinflammatory activity was observed in patient with rheumatoid arthritis (RA) 3 years later. Since then methotrexate established its sound clinical role among cytotoxic antitumour drugs [2]. In adults, MTX is currently used as an anticancer drug in acute lymphoblastic leukemia (ALL) and lymphomas, leukemic and lymphoma CNS involvement, choriocarcinoma, trophoblastic diseases and osteosarcoma. Acute lymphoblastic leukemia (ALL), lymphomas and osteosarcoma are main indications for MTX in pediatric oncological patients. Methotrexate is used as a conventional immunosuppressive drug in rheumatoid arthritis (adult and juvenile) and in psoriasis patients [3].

New discoveries in mechanisms of action

Methotrexate is a competitive inhibitor of dihydrofolatereductase (DHFR), a key enzyme of the folic acid cycle which helps to maintain the intracellular pool of reduced bioactive folates, i.e. the cofactors in one-carbon metabolic pathways [4].

In autoimmune disorders, other mechanisms which suppress immunocompetent and inflammatory cells may predominate including inhibition of enzymes involved in purine synthesis and an augmented release of adenosine as extensively reviewed elsewhere [5, 6]. Antigen-stimulated proliferation of T cells is decreased by methotrexate in vivo by various mechanisms [7, 8]. In polymorfonuclear leukocytes, methotrexate inhibits activity of 5-lipoxygenase and production of leukotriene B4 which acts as a potent inflammation inducer [9–11]. Decreased rate of polyamine synthesis resulting from diminished methyl transfer in cells is another possible cellular mechanism of action [12, 13].

Recently, research has focused on other „non-DHFR related“ mechanisms of action. MTX induced activation of Jun N-terminal kinase (JNK) results in expression of JNK-dependent genes that participate in apoptosis [14, 15]. Downregulation of B cell CLL/lymphoma 2 gene (BCL-2) was observed in leukemic and stomach cancer cell lines treated with methotrexate [16, 17].

Intracellular metabolism

Methotrexate enters the cells through a reduced folate carrier and folate transporters. Other transporters influence the pharmacokinetics of MTX such as the efflux pumps of the ABC superfamily transporters and organic anion transporting peptides [18, 19]. Similarly to folates, MTX undergoes polyglutamation by folylpolyglutamate synthetase (FPGS) [20]. MTX polyglutamates (MTX-Glu2-7) can be viewed as a depot active form of the drug. They are retained intracellularly and slowly leave the cell after enzymatic hydrolysis to parent drug by folylpolyglutamate hydrolase. MTX polyglutamates have much longer half-life (2–3 weeks) as compared to MTX (4–8 hours). Thus, polyglutamation helps to maintain cytotoxic or immunosuppresive concentrations and makes possible intermittent dosing of MTX once a week in the maintenance therapy of ALL or immunosuppressive treatments. There have been several reports that polyglutamation increases variability in methotrexate efficacy [21, 22]. In breast cancer cells, MTX-Glu4-5 showed more prolonged action on DHFR than shorter forms [21]. In addition MTX-Glu2 has been recently proposed as a potential biomarker for clinical response in patients with rheumatoid arthritis treated with methotrexate [23]. Having newly developed methods available that are able to quantitate levels of methotrexate intracellular forms, we expect further elucidation on this topic in the near future [24, 25].

Dosing regimens and effects of extracellular metabolism

MTX dosing vastly varies within clinical protocols for specific diagnoses. Low-dose methotrexate is used to treat RA, psoriasis, and as a maintenance therapy of ALL. According to recommendations of the Czech Society for Rheumatology, low-dose MTX treatment of RA should be initiated at 10–15 mg (oral) weekly [26]. If the clinical response is insufficient, oral administration should be switched for subcutaneous application or oral doses should be increased to 25–30 mg weekly [26]. Initial therapeutic dosage in psoriasis therapy is 7.5 mg MTX weekly that can further be escalated to 30 mg weekly [27]. Acute lymphoblastic leukemia maintenance therapy dose can be 20 mg/m2 oral MTX weekly [28, 29]. High-dose MTX is required in cancer treatment. Pediatric ALL protocols include 5000 mg/m2 IV MTX with leucovorine (LV) rescue (usual dose 15 mg/m2) [30]. Osteosarcoma high-dose IV MTX therapeutic dose is 10 000–12 500 mg/m2 with LV rescue [31]. NHL-BFM 95 protocol contains 1000 mg/m2 IV MTX with LV rescue [32]. Intrathecal MTX is administered at 12 mg/m2 in prophylaxis of leukemic CNS disease in ALL and NHL. Target intrathecal concentration of the drug is 1 µmol/l [30, 32]. Liver function tests and complete blood count with white blood cell count are main markers of MTX clinical toxicity [26, 27]. High-dose MTX protocols require therapeutic drug monitoring (TDM).

Aldehyde oxidase (E.C. 1.2.3.1) converts MTX into its metabolite 7-hydroxymethotrexate (7-OH MTX) [33, 34]. This metabolite, compared to MTX, shows 200 fold less efficacy in blocking DHFR [33–35]. Furthermore, concentrations of 7-OH MTX inversely correlate with clinical toxicity [36]. Decreased levels of 7-OH MTX were reported to show higher therapeutic efficacy [33].

Methotrexate and its clinical safety

Adverse effects of MTX mostly depend on a dosing regimen. The advantage of low-dose MTX lies in its favourable risk benefit ratio. Main adverse effects in low-dose MTX RA treatment are gastrointestinal intolerance and myelosuppression [37]. Psoriasis patients receiving low-dose MTX appear to have increased risk of non-alcoholic fatty liver disease and liver fibrosis [38–40].

Cross-sectional study of 140 patients with RA treated with MTX (10 mg weekly) found 27% with adverse effects, mainly with hepatotoxicity (8.6%, elevated ALT) and anemia (5.7%) [41]. Most common side effect in retrospective study of 71 Arabic patients with RA (7.5 mg MTX weekly with folic acid 2.5–5 mg weekly) were gastrointestinal disturbances (31%) followed by CNS symptoms (18%) and hepatotoxicity (14%, minor to major elevation of liver function tests) [42]. Study of 673 rheumatic patients (between 1986 and 1999) from Staffordshire Rheumatology Centre identified gastrointestinal symptoms (10.8%), hematological abnormalities (5.5%) and abnormal liver function tests (5.5%) as the most common reasons for cessation of MTX treatment [43]. Type II diabetes mellitus and obesity seem to augment adverse effects of MTX treatment in psoriasis [38]. New markers of MTX induced hepatotoxicity in psoriasis and psoriatic arthritis were recently tested and reviewed [39, 40]. In addition, our data from outpatient blood pressure monitoring indicate, that patients with RA and hypertension treated with MTX preserve more normal diurnal pattern of systolic blood pressure values (dippers) than patients treated with NSAIDs and/or corticosteroids [44].

High-dose MTX protocols remain a mainstay in the treatment of childhood malignancies and are clearly reflected in higher and more severe toxicity incidence [45]. Methotrexate high-dose protocols have high risk of nephrotoxicity and tubulopathy that may progress to acute renal damage (ARD) and renal failure [46, 47]. Plasmatic levels of MTX, renal function and hydration must be monitored during high-dose treatment [47]. Appropriate hydration, leucovorine administration and urine alkalization to increase MTX and 7-OH MTX solubility are main preventive steps against crystalluria and ARD [47]. Severe renal impairment during high-dose MTX therapy is managed mainly by high-flux dialysis [47]. Another option is to use carboxypeptidase G2 (glucarpidase), recombinant bacterial enzyme that cleaves carboxyl-terminal glutamate residue from folic acid and its analogues, such as MTX, 7-OH MTX and also LV [48]. Glucarpidase (Voraxaze) was approved by FDA in January 2012. It is IV administered in a single dose of 50 U/kg, however patients should not receive LV within 2 hours before and after administration [47, 49]. Emergency treatment with IT glucarpidase (2000 U reconstituted in 12 ml of normal saline) and systemic LV (4 doses, 100 mg every 6 hours) showed positive outcome in pediatric patients [50]. Therefore IT administration of glucarpidase was proposed as an alternative to standard procedures in intrathecal MTX overdose (drainage by lumbar puncture, ventriculolumbar CSF perfusion, systemic corticosteroids and LV administration) [50].

Allergic reactions to high-dose MTX in children have been reported by various studies although their occurence is relatively rare [51–53]. Another complication in HD MTX treatment is therapy-related neurotoxicity [54]. The authors from the Department of Pediatric Oncology, University Hospital, Brno, described a case of neurotoxicity in 9-year-old boy treated with HD MTX (ALL-BFM 95 protocol). Severe encefalopathy that followed the first HD MTX course in this case correlated with patients five times elevated homocysteine levels. However, these complications, together with laboratory abnormalities, disappeared in the second HD MTX course [55]. Similar cases were observed in other studies [56, 57]. In light of these findings, we may conclude that homocysteine may be a promising pharmacodynamic biomarker of HD MTX-related neurotoxicity in children with cancer.

Conclusion

Methotrexate clinical role within a wide range of modern antineoplastic and immunosuppresive drugs remains untouched. Even though new antifolates are in clinical trials, none of them has shown better risk benefit ratio than the original MTX molecule in terms of pharmacokinetics and pharmacodynamics [58]. High-dose MTX therapeutic protocols went through many years of optimalization and provide safe, efficacious and flexible chemotherapy. We believe that further investigation of MTX effects on cells and finding new but relevant biomarkers in folate metabolism may help consolidate MTX role in current therapeutic protocols and confirm its value in cancer and autoimmunity treatment.

Acknowledgement

All authors listed contributed sufficiently to the manuscript to be included as authors. This work was supported by European Regional Development Fund and the State budget of the Czech Republic for Regional Centre of Applied Molecular Oncology (RECAMO, CZ.1.05/2.1.00/03.0101), by Large Infrastructure Projects of Czech Ministry of Education, Youth and Sports LM2011017 and by IGA Czech Ministry of Health NT14327.

Došlo: 20. 12. 2013

Přijato: 9. 4. 2014

Corresponding author

Bc. Michal Řiháček

Masaryk Memorial Cancer Institute

RECAMO

Žlutý kopec 7

656 53 Brno

Czech Republic

e-mail: michal.rihacek@mou.cz


Sources

1. Farber S, Diamond LK. Temporary remissions in acute leukemia in children produced by folic acid, 4-Aminopteroyl-Glutamic Acid (Aminopterin). N Engl J Med 1948; 23: 787–793.

2. Ward JR. Historical perspective on the use of methotrexate for the treatment of rheumatoid arthritis. J Rheumatol 1985; Suppl: 3–6.

3. Drugs.com. Methotrexate. http://www.drugs.com/methotrexate.html. Accessed January 31, 2014.

4. Valik D, Radina M, Sterba J, et al. Homocysteine: Exploring its potential as a pharmacodynamic biomarker of antifolate chemotherapy. Pharmacogenomics 2004; 8: 1151–1162.

5. Chan ES, Cronstein BN. Methotrexate – how does it really work? Nat Rev Rheumatol 2010; 3: 175–178.

6. Cutolo M, Sulli A, Pizzorni C, et al. Anti-inflammatory mechanisms of methotrexate in rheumatoid arthritis. Ann Rheum Dis 2001; 8: 729–735.

7. Genestier L, Paillot R, Fournel S, et al. Immu-nosuppressive properties of methotrexate: Apoptosis and clonal deletion of activated peripheral t cells. J Clin Invest 1998; 2: 322–328.

8. Johnston A, Gudjonsson JE, Sigmundsdottir H, et al. The anti-inflammatory action of methotrexate is not mediated by lymphocyte apoptosis, but by the suppression of activation and adhesion molecules. Clin Immunol 2005; 2: 154–163.

9. Sperling RI, Benincaso AI, Anderson RJ, et al. Acute and chronic suppression of leukotriene b4 synthesis ex vivo in neutrophils from patients with rheumatoid arthritis beginning treatment with methotrexate. Arthritis Rheum 1992; 4: 376–384.

10. Lammers AM, van de Kerkhof PC, Mier PD. Reduction of leukotriene b4-induced intraepidermal accumulation of polymorphonuclear leukocytes by methotrexate in psoriasis. Br J Dermatol 1987; 5: 667–671.

11. Crooks SW, Stockley RA. Leukotriene b4. Int J Bio-chem Cell Biol 1998; 2: 173–178.

12. Nesher G, Osborn TG, Moore TL. In vitro effects of methotrexate on polyamine levels in lymphocytes from rheumatoid arthritis patients. Clin Exp Rheumatol 1996; 4: 395–399.

13. Nesher G, Osborn TG, Moore TL. Effect of treatment with methotrexate, hydroxychloroquine, and prednisone on lymphocyte polyamine levels in rheumatoid arthritis: Correlation with the clinical response and rheumatoid factor synthesis. Clin Exp Rheumatol 1997; 4: 343–347.

14. Spurlock CF 3rd, Aune ZT, Tossberg JT, et al. Increased sensitivity to apoptosis induced by methotrexate is mediated by jnk. Arthritis Rheum 2011; 9: 2606–2616.

15. Cho HW, Park SK, Heo KW, et al. Methotrexate induces apoptosis in nasal polyps via caspase cascades and both mitochondria-mediated and p38 mitogen-activated protein kinases/jun n-terminal kinase pathways. Am J Rhinol Allergy 2013; 1: e26–31.

16. Florou D, Patsis C, Ardavanis A, et al. Effect of doxorubicin, oxaliplatin, and methotrexate administration on the transcriptional activity of bcl-2 family gene members in stomach cancer cells. Cancer Biol Ther 2013; 7: 587–596.

17. Floros KV, Talieri M, Scorilas A. Topotecan and methotrexate alter expression of the apoptosis-related genes bcl2, fas and bcl2l12 in leukemic hl-60 cells. Biol Chem 2006; 12: 1629–1633.

18. Trevino LR, Shimasaki N, Yang W, et al. Germline genetic variation in an organic anion transporter polypeptide associated with methotrexate pharmacokinetics and clinical effects. J Clin Oncol 2009; 35: 5972–5978.

19. Rau T, Erney B, Gores R, et al. High-dose methotrexate in pediatric acute lymphoblastic leukemia: Impact of abcc2 polymorphisms on plasma concentrations. Clin Pharmacol Ther 2006; 5: 468–476.

20. Westerhof GR, Schornagel JH, Kathmann I, et al. Carrier- and receptor-mediated transport of folate antagonists targeting folate-dependent enzymes: Correlates of molecular-structure and biological activity. Mol Pharmacol 1995; 3: 459–471.

21. Jolivet J, Chabner BA. Intracellular pharmacokinetics of methotrexate polyglutamates in human breast cancer cells. Selective retention and less dissociable binding of 4-nh2-10-ch3-pteroylglutamate4 and 4-nh2-10-ch3-pteroylglutamate5 to dihydrofolate reductase. J Clin Invest 1983; 3: 773–778.

22. Chabner BA, Allegra CJ, Curt GA, et al. Poly-glutamation of methotrexate. Is methotrexate a prodrug? J Clin Invest 1985; 3: 907–912.

23. Hobl EL, Jilma B, Erlacher L, et al. A short-chain methotrexate polyglutamate as outcome parameter in rheumatoid arthritis patients receiving methotrexate. Clin Exp Rheumatol 2012; 2: 156–163.

24. den Boer E, Meesters RJ, van Zelst BD, et al. Measuring methotrexate polyglutamates in red blood cells: A new lc-ms/ms-based method. Anal Bioanal Chem 2013; 5: 1673–1681.

25. Li H, Luo W, Zeng Q, et al. Method for the determination of blood methotrexate by high performance liquid chromatography with online post-column electrochemical oxidation and fluorescence detection. J Chromatogr B Analyt Technol Biomed Life Sci 2007; 1: 164–168.

26. Pavelka K, Venclovsky J. Doporučení české revmatologické společnosti pro léčbu revmatoidní artritidy. Čes Revmatol 2010; 4: 182–191.

27. Farmakoterapie psoriázy. Farmakoterap Inform 2011; 2: 1–3.

28. Schmiegelow K, Bjork O, Glomstein A, et al. Intensification of mercaptopurine/methotrexate maintenance chemotherapy may increase the risk of relapse for some children with acute lymphoblastic leukemia. J Clin Oncol 2003; 7: 1332–1339.

29. Schmiegelow K, Heyman M, Kristinsson J, et al. Oral methotrexate/6-mercaptopurine may be superior to a multidrug lsa2l2 maintenance therapy for higher risk childhood acute lymphoblastic leukemia: Results from the nopho all-92 study. J Pediatr Hematol Oncol 2009; 6: 385–392.

30. Moricke A, Reiter A, Zimmermann M, et al. Risk-adjusted therapy of acute lymphoblastic leukemia can decrease treatment burden and improve survival: Treatment results of 2169 unselected pediatric and adolescent patients enrolled in the trial all-bfm 95. Blood 2008; 9: 4477–4489.

31. Jaffe N, Gorlick R. High-dose methotrexate in osteosarcoma: Let the questions surcease--time for final acceptance. J Clin Oncol 2008; 27: 4365–4366.

32. Woessmann W, Seidemann K, Mann G, et al. The impact of the methotrexate administration schedule and dose in the treatment of children and adolescents with b-cell neoplasms: A report of the bfm group study nhl-bfm95. Blood 2005; 3: 948–958.

33. Baggott JE, Morgan SL. Methotrexate catabolism to 7-hydroxymethotrexate in rheumatoid arthritis alters drug efficacy and retention and is reduced by folic acid supplementation. Arthritis Rheum 2009; 8: 2257–2261.

34. Fabre G, Seither R, Goldman ID. Hydroxylation of 4-amino-antifolates by partially purified aldehyde oxidase from rabbit liver. Biochem Pharmacol 1986; 8: 1325–1330.

35. Farquhar D, Loo TL. Synthesis and biologic evaluation of 7-hydroxymethotrexate, 7-methylaminopterin, and 7-methylmethotrexate. J Med Chem 1972; 5: 567–569.

36. Erttmann R, Bielack S, Landbeck G. 7-hydroxy-methotrexate and clinical toxicity following high-dose methotrexate therapy. J Cancer Res Clin Oncol 1985; 1: 86–88.

37. Schnabel A, Gross WL. Low-dose methotrexate in rheumatic diseases--efficacy, side effects, and risk factors for side effects. Semin Arthritis Rheum 1994; 5: 310–327.

38. Montaudie H, Sbidian E, Paul C, et al. Methotrexate in psoriasis: A systematic review of treatment modalities, incidence, risk factors and monitoring of liver toxicity. J Eur Acad Dermatol Venereol 2011; 12–18.

39. Chladek J, Simkova M, Vaneckova J, et al. Assessment of methotrexate hepatotoxicity in psoriasis patients: A prospective evaluation of four serum fibrosis markers. J Eur Acad Dermatol Venereol 2013; 8: 1007–1014.

40. Lindsay K, Fraser AD, Layton A, et al. Liver fibrosis in patients with psoriasis and psoriatic arthritis on long-term, high cumulative dose methotrexate therapy. Rheumatology (Oxford) 2009; 5: 569–572.

41. Gilani ST, Khan DA, Khan FA, et al. Adverse effects of low dose methotrexate in rheumatoid arthritis patients. J Coll Physicians Surg Pak 2012; 2: 101–104.

42. Attar SM. Adverse effects of low dose methotrexate in rheumatoid arthritis patients. Saudi Med J 2010; 8: 909–915.

43. Kinder AJ, Hassell AB, Brand J, et al. The treatment of inflammatory arthritis with methotrexate in clinical practice: Treatment duration and incidence of adverse drug reactions. Rheumatology (Oxford) 2005; 1: 61–66.

44. Rihacek I, Frana P, Soucek M, et al. The diurnal variability of blood pressure in patients with hypertension and rheumatoid arthritis. Vnitr Lek 2009; 2: 111–116.

45. Sterba J, Valik D, Bajciova V, et al. High-dose methotrexate and/or leucovorin rescue for the treatment of children with lymphoblastic malignancies: Do we really know why, when and how? Neoplasma 2005; 6: 456–463.

46. Widemann BC, Adamson PC. Understanding and managing methotrexate nephrotoxicity. Oncologist 2006; 6: 694–703.

47. Rysava R. Glomerular impairment associated with malignancies and chemotherapy induced nephrotoxicity. Postgrad Nefrol 2013; 4: 50–53.

48. Schwartz S, Borner K, Muller K, et al. Glucarpidase (carboxypeptidase g2) intervention in adult and elderly cancer patients with renal dysfunction and delayed methotrexate elimination after high-dose methotrexate therapy. Oncologist 2007; 11: 1299–1308.

49. Green JM. Glucarpidase to combat toxic levels of methotrexate in patients. Ther Clin Risk Manag 2012; 403–413.

50. Widemann BC, Balis FM, Shalabi A, et al. Treatment of accidental intrathecal methotrexate overdose with intrathecal carboxypeptidase g2. J Natl Cancer Inst 2004; 20: 1557–1559.

51. Oulego-Erroz I, Maneiro-Freire M, Bouzon-Alejandro M, et al. Anaphylactoid reaction to high-dose methotrexate and successful desensitization. Pediatr Blood Cancer 2010; 3: 557–559.

52. Umeda T, Takada N, Hodaka E, et al. [evaluation of severe side effects of high-dose methotrexate in osteosarcoma]. Gan To Kagaku Ryoho 1984; 2: 285–294.

53. Postovsky S, Elhasid R, Ben-Barak A, et al. Allergic reaction to high-dose methotrexate. Med Pediatr Oncol 2000; 2: 131–132.

54. Drachtman RA, Cole PD, Golden CB, et al. Dextromethorphan is effective in the treatment of subacute methotrexate. Pediatr Hematol Oncol 2002; 5: 319–327.

55. Valik D, Sterba J, Bajciova V, et al. Severe encephalopathy induced by the first but not the second course of high-dose methotrexate mirrored by plasma homocysteine elevations and preceded by extreme differences in pretreatment plasma folate. Oncology 2005; 3: 269–272.

56. Quinn CT, Griener JC, Bottiglieri T, et al. Elevation of homocysteine and excitatory amino acid neurotransmitters in the csf of children who receive methotrexate for the treatment of cancer. J Clin Oncol 1997; 8: 2800–2806.

57. Becker A, Vezmar S, Linnebank M, et al. Marked elevation in homocysteine and homocysteine sulfinic acid in the cerebrospinal fluid of lymphoma patients receiving intensive treatment with methotrexate. Int J Clin Pharmacol Ther 2007; 9: 504–515.

58. Takimoto CH. New antifolates: Pharmacology and clinical applications. Oncologist 1996; 1 & 2: 68–81.

Labels
Neonatology Paediatrics General practitioner for children and adolescents
Login
Forgotten password

Enter the email address that you registered with. We will send you instructions on how to set a new password.

Login

Don‘t have an account?  Create new account

#ADS_BOTTOM_SCRIPTS#