Zeitschrift für Komplementärmedizin 2018; 10(03): 50-57
DOI: 10.1055/a-0625-7414
Praxis
Kampo-Medizin
© Karl F. Haug Verlag in MVS Medizinverlage Stuttgart GmbH & Co. KG

Kampo-Medizin in der gastrointestinalen Onkologie

Silke Cameron

Subject Editor:
Further Information

Publication History

Publication Date:
11 June 2018 (online)

Summary

In Japan werden Rezepturen der traditionellen Phytotherapie (Kampo-Medizin) seit langem im modernen Gesundheitssystem zur komplementären Behandlung in der Onkologie evidenzbasiert eingesetzt. Die Kampo-Medizin bietet ein großes Spektrum für die Behandlung von Tumorpatienten: Um Symptome durch die Tumorerkrankung selbst mildern, Nebenwirkungen von Chemotherapeutika oder Radiotherapien zu verbessern. Auch postoperative Motilitätsstörungen oder die Opiat-induzierte Koprostase können behandelt werden. Insbesondere in der palliativen Medizin, aber auch bei älteren Patienten, bei denen ein Polypharmazie-­Effekt verhindert werden sollte, leistet die Kampo-Medizin wertvolle Dienste.

In diesem Artikel werden wichtige Kampo-Rezepturen für die supportive Tumorbehandlung bei häufigen Indikationen vorgestellt sowie der aktuelle japanische Forschungsstand referiert.

 
  • Literatur

  • 1 Reissenweber-Hewel H. Japanische Kampo-Medizin. Pharmakon 2014; 127: 134
  • 2 Ito A, Munakata K, Imazu Y. et al. First nationwide attitude survey of Japanese physicians on the use of traditional Japanese medicine (kampo) in cancer treatment. Evid Based Complement Alternat Med 2012; 2012: 957-082
  • 3 Eberhard U. Leitfaden Kampo Medizin – Japanische Phytotherapie. München, Jena; Elsevier: 2003
  • 4 Ishimaru N, Maeno T, Suzuki M. et al. Rapid effects of Kikyo-to on sore throat pain associated with acute upper respiratory tract infection. J Complement Integr Med 2013; 11: 51-54
  • 5 Tanemoto R, Okuyama T, Matsuo H. et al. The constituents of licorice (Glycyrrhiza uralensis) differentially suppress nitric oxide production in interleukin-1beta-treated hepatocytes. Biochem Biophys Rep 2015; 2: 153-159
  • 6 Langer D, Czarczynska-Goslinska B, Goslinski T. Glycyrrhetinic acid and its derivatives in infectious diseases. Current Issues in Pharmacy and Medical Sciences. 2016 doi:10.1515/cipms-2016–0024
  • 7 Ozaki Y. Studies on antiinflammatory effect of Japanese Oriental medicines (kampo medicines) used to treat inflammatory diseases. Biol Pharm Bull 1995; 18: 559-562
  • 8 WHO Monographs on Selected Medicinal Plants Volume 1:, 1999
  • 9 Katagiri F, Itoh H, Takeyama M. Effect of Sho-hange-ka-bukuryo-to on gastrointestinal peptide concentrations in the plasma of healthy human subjects. Biol Pharm Bull 2004; 27: 1674-1678
  • 10 Saitoh K, Kase Y, Ishige A. et al. Effects of Keishi-ka-shakuyaku-to (Gui-Zhi-Jia-Shao-Yao-Tang) on diarrhea and small intestinal movement. Biol Pharm Bull 1999; 22: 87-89
  • 11 Ahrens T. Kampo in der Gastroenterologie. Praktikable Konzepte für die integrative Therapie von Beschwerden des Gastrointestinaltraktes mit traditioneller japanischer Phytotherapie. Z Phytother 2017; 38 (06) 260-265
  • 12 Mori K, Kondo T, Kamiyama Y. Preventive effect of Kampo medicine (Hangeshashin-to) against irinotecan-induced diarrhea in advanced non-small-cell lung cancer. Cancer Chemother Pharmacol 2003; 51: 403-406
  • 13 Mathijssen RH, van Alphen RJ, Verweij J. et al. Clinical pharmacokinetics and metabolism of irinotecan (CPT-11). Clin Cancer Res 2001; 7: 2182-2194
  • 14 Narita M, Nagai E, Hagiwara H. et al. Inhibition of beta-glucuronidase by natural glucuronides of kampo medicines using glucuronide of SN-38 (7-ethyl-10-hydroxycamptothecin) as a substrate. Xenobiotica 1993; 23: 5-10
  • 15 Takasuna K, Kasai Y, Kitano Y. et al. Protective effects of kampo medicines and baicalin against intestinal toxicity of a new anticancer camptothecin derivative, irinotecan hydrochloride (CPT-11), in rats. Jpn J Cancer Res 1995; 86: 978-984
  • 16 Kase Y, Hayakawa T, Ishige A. et al. The effects of Hange-shashin-to on the content of prostaglandin E2 and water absorption in the large intestine of rats. Biol Pharm Bull 1997; 20: 954-957
  • 17 Sharma R, Tobin P, Clarke SJ. Management of chemotherapy-induced nausea, vomiting, oral mucositis, and diarrhoea. Lancet Oncol 2005; 6: 93-102
  • 18 Suzuki H, Matsuzaki J, Fukushima Y. et al. Randomized clinical trial: rikkunshito in the treatment of functional dyspepsia--a multicenter, double-blind, randomized, placebo-controlled study. Neurogastroenterol Motil 2014; 26: 950-961
  • 19 Takahashi T, Endo S, Nakajima K. et al. Effect of rikkunshito, a chinese herbal medicine, on stasis in patients after pylorus-preserving gastrectomy. World J Surg 2009; 33: 296-302
  • 20 Tominaga K, Kido T, Ochi M. et al. The traditional Japanese medicine Rikkunshito promotes gastric emptying via the antagonistic action of the 5-HT(3) receptor pathway in rats. Evid Based Complement Alternat Med 2011; 2011: 248-481
  • 21 Tominaga K, Arakawa T. Kampo medicines for gastrointestinal tract disorders: a review of basic science and clinical evidence and their future application. J Gastroenterol 2013; 48: 452-462
  • 22 Nahata M, Saegusa Y, Sadakane C. et al. Administration of exogenous acylated ghrelin or rikkunshito, an endogenous ghrelin enhancer, improves the decrease in postprandial gastric motility in an acute restraint stress mouse model. Neurogastroenterol Motil 2014; 26: 821-831
  • 23 Goso Y, Ogata Y, Ishihara K. et al. Effects of traditional herbal medicine on gastric mucin against ethanol-induced gastric injury in rats. Comp Biochem Physiol C Pharmacol Toxicol Endocrinol 1996; 113: 17-21
  • 24 Shiratori M, Shoji T. et al. Effect of rikkunshito on gastric sensorimotor function under distention. Neurogastroenterol Motil 2011; 23: 323-329
  • 25 Ohno T, Yanai M, Ando H. et al. Rikkunshito, a traditional Japanese medicine, suppresses cisplatin-induced anorexia in humans. Clin Exp Gastroenterol 2011; 4: 291-296
  • 26 Seike J, Sawada T, Kawakita N. et al. A new candidate supporting drug, Rikkunshito, for the QOL in advanced esophageal cancer patients with chemotherapy using docetaxel/5-FU / CDDP. Int J Surg Oncol 2011; 2011: 715-623
  • 27 Miura N, Fukutake M, Yamamoto M. et al. An herbal medicine orengedokuto prevents indomethacin-induced enteropathy. Biol Pharm Bull 2007; 30: 495-501
  • 28 Nishioka M, Shimada M, Kurita N. et al. The Kampo medicine, Goshajinkigan, prevents neuropathy in patients treated by FOLFOX regimen. Int J Clin Oncol 2011; 16: 322-327
  • 29 Kono T, Hata T, Morita S. et al. Goshajinkigan oxaliplatin neurotoxicity evaluation (GONE): a phase 2, multicenter, randomized, doubleblind, placebocontrolled trial of goshajinkigan to prevent oxaliplatininduced neuropathy. Cancer Chemother Pharmacol 2013; 72: 1283-1290
  • 30 Nishizawa M, Sutherland WH, Nukada H. Gosha-jinki-gan (herbal medicine) in streptozocin-induced diabetic neuropathy. J Neurol Sci 1995; 132: 177-181
  • 31 Reissenweber H, Cameron S, Landgraf R. Clinical Evaluation of Japanese Phytotherapy (Gosha-jinki-gan) in Diabetic Neuropathy: A Prospective Open Pilot Study. Abstract for the 6th International ESCOP Symposium: Herbal medicinal products -scientific strategies in Europe.
  • 32 Cameron-Schaefer S, Kondo K, Ishige A. et al. Maintaining the redox-balance intact: gosha-jinki-gan but not insulin activates retinal soluble guanylate cyclase in diabetic rats. Ophthalmic Res 2006; 38: 95-104
  • 33 Kito Y, Teramoto N. Effects of Hange-shashin-to (TJ-14) and Keishi-ka-shakuyaku-to (TJ-60) on contractile activity of circular smooth muscle of the rat distal colon. Am J Physiol Gastrointest Liver Physiol 2012; 303: G1059-G1066
  • 34 Ai M, Yamaguchi T, Odaka T. et al. Objective assessment of the antispasmodic effect of shakuyaku-kanzo-to (TJ-68), a Chinese herbal medicine, on the colonic wall by direct spraying during colonoscopy. World J Gastroenterol 2006; 12: 760-764
  • 35 Sakai Y, Tsuyuguchi T, Ishihara T. et al. Confirmation of the antispasmodic effect of shakuyaku-kanzo-to (TJ-68), a Chinese herbal medicine, on the duodenal wall by direct spraying during endoscopic retrograde cholangiopancreatography. J Nat Med 2009; 63: 200-203
  • 36 Yoshida T, Sawa T, Ishiguro T. et al. The efficacy of prophylactic Shakuyaku-Kanzo-to for myalgia and arthralgia following carboplatin and paclitaxel combination chemotherapy for non-small cell lung cancer. Support Care Cancer 2009; 17: 315-320
  • 37 Kuchta K. 3 Fachleute–3 Behandlungsstrategien: Lebensverlängerndes Phytotherapeutikum weiterhin gesucht! Zkm 2015; 6: 40-41
  • 38 Jeong JS, Ryu BH, Kim JS. et al. Bojungikki-tang for cancer-related fatigue: a pilot randomized clinical trial. Integr Cancer Ther 2010; 9: 331-338
  • 39 Yae S, Takahashi F, Yae T. et al. Hochuekkito (TJ-41), a Kampo formula, ameliorates cachexia induced by colon 26 adenocarcinoma in mice. Evid Based Complement Alternat Med 2012; 2012: 976-926
  • 40 Cho JM, Sato N, Kikuchi K. Prophylactic anti-tumor effect of Hochu-ekki-to (TJ41) by enhancing natural killer cell activity. In Vivo 1991; 5: 389-391
  • 41 Sato T, Kita K, Sato C. et al. Hochuekkito (Buzhongyiqitang), a herbal medicine, enhances cisplatininduced apoptosis in HeLa cells. Mol Med Rep 2015; 12: 6215-6220
  • 42 Yu N, Xiong Y, Wang C. Bu-Zhong-Yi-Qi decoction, the water extract of Chinese traditional herbal medicine, enhances cisplatin cytotoxicity in A549/DDP cells through induction of apoptosis and autophagy. Biomed Res Int 2017; 2017: 3692797
  • 43 Ikemoto T, Shimada M, Iwahashi S. et al. Changes of immunological parameters with administration of Japanese Kampo medicine (Juzen-Taihoto / TJ-48) in patients with advanced pancreatic cancer. Int J Clin Oncol 2014; 19: 81-86
  • 44 Ogawa K, Omatsu T, Matsumoto C. et al. Protective effect of the Japanese traditional medicine juzentaihoto on myelosuppression induced by the anticancer drug TS-1 and identification of a potential biomarker of this effect. BMC Complement Altern Med 2012; 12: 118
  • 45 Matsuda T, Maekawa K, Asano K. et al. Suppressive effect of juzen-taiho-to on lung metastasis of b16 melanoma cells in vivo. Evid Based Complement Alternat Med 2011; 2011: 743-153
  • 46 Okumi H, Koyama A. Kampo medicine for palliative care in Japan. Biopsychosoc Med 2014; 8: 6
  • 47 Ohnishi Y, Fujii H, Hayakawa Y. et al. Oral administration of a Kampo (Japanese herbal) medicine Juzen-taiho-to inhibits liver metastasis of colon 26-L5 carcinoma cells. Jpn J Cancer Res 1998; 89: 206-213
  • 48 Yaegashi M, Otsuka K, Itabashi T. et al. Daikenchuto stimulates colonic motility after laparoscopic-assisted colectomy. Hepatogastroenterology 2014; 61: 85-89
  • 49 Fukuda H, Chen C, Mantyh C. et al. The herbal medicine, Dai-Kenchu-to, accelerates delayed gastrointestinal transit after the operation in rats. J Surg Res 2006; 131: 290-295
  • 50 Akamaru Y, Takahashi T, Nishida T. et al. Effects of daikenchuto, a Japanese herb, on intestinal motility after total gastrectomy: a prospective randomized trial. J Gastrointest Surg 2015; 19: 467-472
  • 51 Yamada T, Matsumoto S, Matsuda MKA. et al. The effect of Daikenchuto on postoperative intestinal motility in patients with right-side colon cancer. Surg Today 2017; 47: 865-871
  • 52 Fukuda H, Chen C, Mantyh C. et al. The herbal medicine, Dai-Kenchu-to, accelerates delayed gastrointestinal transit after the operation in rats. J Surg Res 2006; 131: 290-295
  • 53 Nakamura T, Sakai A, Isogami I. et al. Abatement of morphine-induced slowing in gastrointestinal transit by Dai-kenchu-to, a traditional Japanese herbal medicine. Jpn J Pharmacol 2002; 88: 217-221
  • 54 Takayama S, Seki T, Watanabe M. et al. The herbal medicine Daikenchuto increases blood flow in the superior mesenteric artery. Tohoku J Exp Med 2009; 219: 319-330
  • 55 Kono T, Kanematsu T, Kitajima M. Exodus of Kampo, traditional Japanese medicine, from the complementary and alternative medicines: is it time yet?. Surgery 2009; 146: 837-840
  • 56 Okada K, Shoda J, Kano M. et al. Inchinkoto, a herbal medicine, and its ingredients dually exert Mrp2/MRP2-mediated choleresis and Nrf2-mediated antioxidative action in rat livers. Am J Physiol Gastrointest Liver Physiol 2007; 292: G1450-G1463
  • 57 Evers R, de HM, Sparidans R, Beijnen J. et al. Vinblastine and sulfinpyrazone export by the multidrug resistance protein MRP2 is associated with glutathione export. Br J Cancer 2000; 83: 375-383