Preclinical in vivo

Explore the preclinical in vivo studies of kratom and the active compounds extracted from the plant, alkaloids.

To determine the safety and efficacy of any compound, preclinical in vivo testing in mammalian species must be performed. For a compound to become recognized as safe, approved as a dietary supplement, or sold as a drug product (over the counter or prescription) it must undergo a variety of studies to ensure its long-term safe use and recommended use levels. When it comes to products derived from botanicals or botanicals themselves, testing becomes even more complicated, as plants may contain numerous bioactive compounds. 

Testing is performed in rodents first (mice or rats) prior to being carried out in larger nonrodent species (dogs, monkeys). The following will explore the preclinical work performed for kratom and its isolated alkaloids.

Preclinical in vivo receptor pharmacology, efficacy, and abuse potential

Receptor Pharmacology

In preclinical in vivo research, rodent and nonrodent models are used to determine safety and efficacy of compounds. In rodent models, rats and mice are used to determine the mechanism of action of a compound using various methods. For opioid receptor studies, hot-plate assays or other measures of analgesia are used to determine if a compound is acting with the central opioid system. The compound is measured against a gold-standard compound (for these studies usually morphine) and an antagonist is used to ensure that the mechanism of action is through the receptor system being explored.

VIDEO: In Vivo Receptor Pharmacology

Kratom extracts, as well as isolated mitragynine, show central analgesia in rodents which is fully antagonized by non-selective opioid antagonists (i.e., naloxone or naltrexone) suggesting that analgesia from kratom is primarily driven by the opioid receptor system (1-4). Other isolated alkaloids have been tested with speciociliatine and corynoxine, two other minor indole alkaloids found in kratom, having equal or more potency than morphine (5). Another kratom alkaloid, corynantheidine was found to be an antagonist at opioid receptors (6). Mitragynine has also been found to act through central adrenergic analgesic pathways in rodents indicating its pain relieving/reducing properties occur through activation of multiple receptor systems (7, 8). 

As kratom is also used traditionally to increase mood, activity of kratom and its alkaloids at serotonin and dopamine receptors has been researched in vivo. Serotonin receptors play an important role in regulation of mood, cognition, and sleep, among other physiological processes. A measure of in vivo serotonin activity in rodents is the lower lip retraction test. In this test, the alkaloids paynantheine and speciogynine demonstrated the greatest activity and this activity was antagonized by WAY100635, a serotonin receptor antagonist, so both alkaloids act as either agonists or partial agonists at serotonin receptors (9). Dopamine receptors are important for regulation of emotion, learning, and play a role in reward systems among other functions and have been studied in rodent models with kratom extract and isolated mitragynine. Mitragynine shows anxiolytic properties in rodents that are reversed by opioid, GABA, and dopamine antagonists which indicates that mitragynine may decrease anxiety through a variety of receptor subtypes, as it does with analgesia (10, Hazim, 2011 #593, 11) On the other hand, kratom methanolic extracts have shown an interesting duality when tested in rodent models of psychosis. In certain instances, kratom exacerbated symptoms of psychosis while in other tests it decreased psychosis through the dopaminergic system which underlines the need for more study to understand the dose-response relationship of individual kratom alkaloids and full kratom products (12). 

Efficacy for substance use disorders

VIDEO: Efficacy for substance use disorders

In addition to the preclinical work described above that shows kratom and/or its alkaloids have potential for pain and mental health, research studying kratom and its alkaloids for substance use disorders has been performed. A lyophilized kratom tea preparation significantly decreased opioid withdrawal in morphine dependent mice indicating that full leaf kratom preparations may be useful for opioid withdrawal symptoms. Importantly, the kratom tea preparation did not produce any physical dependence in mice when administered chronically (13).

 In another study, kratom extract and mitragynine were separately investigated to determine if they reduced withdrawal symptoms in morphine dependent mice. Whole kratom extract and mitragynine isolate both decrease withdrawal in opioid dependent subjects and the effect of mitragynine resembled that produced by methadone and buprenorphine, two currently approved medications for opioid use disorder (14, 15). Mitragynine has also demonstrated attenuation of morphine dependence, reduction in response rates of heroin induced conditioned place preference, and reduction of morphine tolerance (16-20).

Kratom extracts and alkaloids have also demonstrated utility in alcohol withdrawal, decreasing alcohol seeking behaviors, and decreasing alcohol intake in rodents (21-24).

Abuse Potential

When the United States Drug Enforcement Agency considers a compound for scheduling as part of the Controlled Substances Act, eight factor analysis is performed. This analysis includes the abuse potential, pharmacology, current scientific knowledge, history and patterns of abuse, significance of abuse, risk to public health, dependence, and comparison to already controlled substances. The abuse potential of kratom and its alkaloids have been assessed preclinically using self-administration, drug discrimination, conditioned place preference, dependence, and withdrawal assays.

VIDEO: Abuse Potential

In intravenous self-administration studies of mitragynine, no abuse potential is signaled and mitragynine pretreatment decreased heroin self-administration (19, 20). But the metabolite of mitragynine, 7-hydroxymitragynine, does show abuse potential when measured against morphine in self-administration procedures (20). In tests of drug-discrimination, mitragynine has the strongest generalization to lofexidine (an α-2 adrenergic receptor agonist used for acute opioid withdrawal symptoms) and phenylephrine (found in many over the counter cold medications as a nasal decongestant) both of which are not controlled substances (25). High doses of kratom methanolic extract achieved conditioned place preference while lower doses of lyophilized kratom tea did not produce conditioned place preference which is a measure of the rewarding properties of a compound (13, 24). Mitragynine causes no spontaneous withdrawal upon cessation, but naloxone precipitated withdrawal symptoms are apparent though weaker and short-lived compared to the morphine control group (26).

Overall, though dependence and withdrawal can be simulated in rodent models, it is modest and manageable compared to other opioids, so the abuse liability of kratom does not warrant it becoming a controlled substance.

Preclinical in vivo pharmacokinetics and safety

Preclinically, short-term or single doses of kratom and individual alkaloids are well tolerated in mice, rats, dogs, and monkeys (27-31), (9, 32-35). Preclinical safety studies in rodent and nonrodent species are lacking for kratom and its alkaloids. No acute or chronic toxicity or carcinogenicity studies have been performed. To better understand the safety profile of kratom, clinical data in humans from long-term users is analyzed.

SOURCES

  1. Shaik Mossadeq WM, Sulaiman MR, Tengku Mohamad TA, Chiong HS, Zakaria ZA, Jabit ML, et al. Anti-inflammatory and antinociceptive effects of Mitragyna speciosa Korth methanolic extract. Medical principles and practice : international journal of the Kuwait University, Health Science Centre. 2009;18(5):378-84. doi: 10.1159/000226292. (Free)

  2. Carpenter JM, Criddle CA, Craig HK, Ali Z, Zhang Z, Khan IA, et al. Comparative effects of Mitragyna speciosa extract, mitragynine, and opioid agonists on thermal nociception in rats. Fitoterapia. 2016;109:87-90. doi: 10.1016/j.fitote.2015.12.001.

  3. Sabetghadam A, Ramanathan S, Mansor SM. The evaluation of antinociceptive activity of alkaloid, methanolic, and aqueous extracts of Malaysian Mitragyna speciosa Korth leaves in rats. Pharmacognosy research. 2010;2(3):181-5. doi: 10.4103/0974-8490.65514. (Free)

  4. Matsumoto K, Mizowaki M, Takayama H, Sakai S, Aimi N, Watanabe H. Suppressive effect of mitragynine on the 5-methoxy-N,N-dimethyltryptamine-induced head-twitch response in mice. Pharmacol Biochem Behav. 1997;57(1-2):319-23. doi: 10.1016/s0091-3057(96)00314-0.

  5. Obeng S, Kamble SH, Reeves ME, Restrepo LF, Patel A, Behnke M, et al. Investigation of the adrenergic and opioid binding affinities, metabolic stability, plasma protein binding properties, and functional effects of selected indole-based kratom alkaloids. J Med Chem. 2020;63(1):433-9. doi: 10.1021/acs.jmedchem.9b01465. (Free)

  6. Takayama H, Ishikawa H, Kurihara M, Kitajima M, Aimi N, Ponglux D, et al. Studies on the synthesis and opioid agonistic activities of mitragynine-related indole alkaloids: discovery of opioid agonists structurally different from other opioid ligands. J Med Chem. 2002;45(9):1949-56. doi: 10.1021/jm010576e.

  7. Matsumoto K, Mizowaki M, Suchitra T, Murakami Y, Takayama H, Sakai S-i, et al. Central antinociceptive effects of mitragynine in mice: contribution of descending noradrenergic and serotonergic systems. European Journal of Pharmacology. 1996;317(1):75-81. doi: https://doi.org/10.1016/S0014-2999(96)00714-5.

  8. Foss JD, Nayak SU, Tallarida CS, Farkas DJ, Ward SJ, Rawls SM. Mitragynine, bioactive alkaloid of kratom, reduces chemotherapy-induced neuropathic pain in rats through α-adrenoceptor mechanism. Drug Alcohol Depend. 2020;209:107946. doi: 10.1016/j.drugalcdep.2020.107946. (Free)

  9. Leon F, Obeng S, Mottinelli M, Chen Y, King TI, Berthold EC, et al. Activity of Mitragyna speciosa ("Kratom") Alkaloids at Serotonin Receptors. J Med Chem. 2021;64(18):13510-23. doi: 10.1021/acs.jmedchem.1c00726. (Free)

  10. Hazim AI, Ramanathan S, Parthasarathy S, Muzaimi M, Mansor SM. Anxiolytic-like effects of mitragynine in the open-field and elevated plus-maze tests in rats. The Journal of Physiological Sciences. 2014;64(3):161-9. doi: 10.1007/s12576-014-0304-0. (Free)

  11. Hazim AI, Mustapha M, Mansor SM. The effects on motor behaviour and short-term memory tasks in mice following an acute administration of Mitragyna speciosa alkaloid extract and mitragynine. Journal of Medicinal Plants Research. 2011;5(24):5810-7. (Free)

  12. Vijeepallam K, Pandy V, Kunasegaran T, Murugan DD, Naidu M. Mitragyna speciosa Leaf Extract Exhibits Antipsychotic-Like Effect with the Potential to Alleviate Positive and Negative Symptoms of Psychosis in Mice. Front Pharmacol. 2016;7:464. doi: 10.3389/fphar.2016.00464. (Free)

  13. Wilson LL, Harris HM, Eans SO, Brice-Tutt AC, Cirino TJ, Stacy HM, et al. Lyophilized Kratom Tea as a Therapeutic Option for Opioid Dependence. Drug Alcohol Depend. 2020;216:108310. doi: 10.1016/j.drugalcdep.2020.108310.

  14. Wilson LL, Chakraborty S, Eans SO, Cirino TJ, Stacy HM, Simons CA, et al. Kratom Alkaloids, Natural and Semi-Synthetic, Show Less Physical Dependence and Ameliorate Opioid Withdrawal. Cellular and molecular neurobiology. 2021;41(5):1131-43. doi: 10.1007/s10571-020-01034-7. (Free)

  15. Hassan R, Pike See C, Sreenivasan S, Mansor SM, Müller CP, Hassan Z. Mitragynine Attenuates Morphine Withdrawal Effects in Rats-A Comparison With Methadone and Buprenorphine. Front Psychiatry. 2020;11:411. doi: 10.3389/fpsyt.2020.00411. (Free)

  16. Meepong R, Sooksawate T. Mitragynine reduced morphine-induced conditioned place preference and withdrawal in rodents. Thai Journal of Pharmaceutical Sciences (TJPS). 2019;43(1). (Fere)

  17. Jamil MF, Subki MF, Lan TM, Majid MI, Adenan MI. The effect of mitragynine on cAMP formation and mRNA expression of mu-opioid receptors mediated by chronic morphine treatment in SK-N-SH neuroblastoma cell. J Ethnopharmacol. 2013;148(1):135-43. doi: 10.1016/j.jep.2013.03.078.

  18. Fakurazi S, Rahman SA, Hidayat MT, Ithnin H, Moklas MA, Arulselvan P. The combination of mitragynine and morphine prevents the development of morphine tolerance in mice. Molecules. 2013;18(1):666-81. doi: 10.3390/molecules18010666. (Free)

  19. Yue K, Kopajtic TA, Katz JL. Abuse liability of mitragynine assessed with a self-administration procedure in rats. Psychopharmacology (Berl). 2018;235(10):2823-9. doi: 10.1007/s00213-018-4974-9.

  20. Hemby SE, McIntosh S, Leon F, Cutler SJ, McCurdy CR. Abuse liability and therapeutic potential of the Mitragyna speciosa (kratom) alkaloids mitragynine and 7-hydroxymitragynine. Addict Biol. 2019;24(5):874-85. doi: 10.1111/adb.12639.

  21. Kumarnsit E, Keawpradub N, Nuankaew W. Effect of Mitragyna speciosa aqueous extract on ethanol withdrawal symptoms in mice. Fitoterapia. 2007;78(3):182-5. doi: https://doi.org/10.1016/j.fitote.2006.11.012.

  22. Cheaha D, Keawpradub N, Sawangjaroen K, Phukpattaranont P, Kumarnsit E. Effects of an alkaloid-rich extract from Mitragyna speciosa leaves and fluoxetine on sleep profiles, EEG spectral frequency and ethanol withdrawal symptoms in rats. Phytomedicine. 2015;22(11):1000-8. doi: https://doi.org/10.1016/j.phymed.2015.07.008.

  23. Gutridge AM, Robins MT, Cassell RJ, Uprety R, Mores KL, Ko MJ, et al. G protein-biased kratom-alkaloids and synthetic carfentanil-amide opioids as potential treatments for alcohol use disorder. British Journal of Pharmacology. 2020;177(7):1497-513. doi: https://doi.org/10.1111/bph.14913. (Free)

  24. Vijeepallam K, Pandy V, Murugan DD, Naidu M. Methanolic extract of Mitragyna speciosa Korth leaf inhibits ethanol seeking behaviour in mice: involvement of antidopaminergic mechanism. Metabolic brain disease. 2019;34(6):1713-22. doi: 10.1007/s11011-019-00477-2.

  25. Reeve ME, Obeng S, Oyola FL, Behnke M, Restrepo LF, Patel A, et al. The Adrenergic a2 Receptor‐Mediated Discriminative‐Stimulus Effects of Mitragynine, the Primary Alkaloid in Kratom (Mitragyna Speciosa) in Rats. The FASEB Journal. 2020;34(S1):1-. (Free)

  26. Harun N, Johari IS, Mansor SM, Shoaib M. Assessing physiological dependence and withdrawal potential of mitragynine using schedule-controlled behaviour in rats. Psychopharmacology (Berl). 2020;237(3):855-67. doi: 10.1007/s00213-019-05418-6.

  27. Ya K, Tangamornsuksan W, Scholfield CN, Methaneethorn J, Lohitnavy M. Pharmacokinetics of mitragynine, a major analgesic alkaloid in kratom (Mitragyna speciosa): A systematic review. Asian Journal of Psychiatry. 2019;43:73-82. doi: https://doi.org/10.1016/j.ajp.2019.05.016.

  28. Kamble SH, Berthold EC, King TI, Raju Kanumuri SR, Popa R, Herting JR, et al. Pharmacokinetics of eleven kratom alkaloids following an oral dose of either traditional or commercial kratom products in rats. J Nat Prod. 2021;84(4):1104-12. doi: 10.1021/acs.jnatprod.0c01163. (Free)

  29. Maxwell EA, King TI, Kamble SH, Raju KSR, Berthold EC, Leon F, et al. Pharmacokinetics and safety of mitragynine in beagle dogs. Planta Med. 2020;86(17):1278-85. doi: 10.1055/a-1212-5475. (Free)

  30. Kong WM, Mohamed Z, Alshawsh MA, Chik Z. Evaluation of pharmacokinetics and blood-brain barrier permeability of mitragynine using in vivo microdialysis technique. J Pharm Biomed Anal. 2017;143:43-7. doi: https://doi.org/10.1016/j.jpba.2017.05.020.

  31. Avery BA, Boddu SP, Sharma A, Furr EB, Leon F, Cutler SJ, et al. Comparative pharmacokinetics of mitragynine after oral administration of Mitragyna speciosa (kratom) leaf extracts in rats. Planta Med. 2019;85(4):340-6. doi: 10.1055/a-0770-3683.

  32. Berthold EC, Kamble SH, Raju KS, King TI, Popa R, Sharma A, et al. Preclinical pharmacokinetic study of speciociliatine, a kratom alkaloid, in rats using an UPLC-MS/MS method. J Pharm Biomed Anal. 2021;194:113778. doi: 10.1016/j.jpba.2020.113778.

  33. Maxwell EA, King TI, Kamble SH, Raju KSR, Berthold EC, Leon F, et al. Oral Pharmacokinetics in Beagle Dogs of the Mitragynine Metabolite, 7-Hydroxymitragynine. Eur J Drug Metab Pharmacokinet. 2021;46(3):459-63. doi: 10.1007/s13318-021-00684-2. (Free)

  34. Berthold EC, Kamble SH, Raju KS, Kuntz MA, Senetra AS, Mottinelli M, et al. The Lack of Contribution of 7-Hydroxymitragynine to the Antinociceptive Effects of Mitragynine in Mice: A Pharmacokinetic and Pharmacodynamic Study. Drug Metab Dispos. 2022;50(2):158-67. doi: 10.1124/dmd.121.000640. (Free)

  35. King TI, Sharma A, Kamble SH, Leon F, Berthold EC, Popa R, et al. Bioanalytical method development and validation of corynantheidine, a kratom alkaloid, using UPLC-MS/MS, and its application to preclinical pharmacokinetic studies. J Pharm Biomed Anal. 2020;180:113019. doi: 10.1016/j.jpba.2019.113019. (Free)