• No results found

University of Groningen Improving antimicrobial therapy for Buruli ulcer Omansen, Till Frederik

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Improving antimicrobial therapy for Buruli ulcer Omansen, Till Frederik"

Copied!
11
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

University of Groningen

Improving antimicrobial therapy for Buruli ulcer

Omansen, Till Frederik

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2019

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Omansen, T. F. (2019). Improving antimicrobial therapy for Buruli ulcer: Pre-clinical studies towards highly efficient, short-course therapy. University of Groningen.

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Chapter 11

discussion and Future perspectives

The future of Buruli ulcer treatment

Manuscript in preparation.

Till F. Omansen1,2, Ymkje Stienstra2, Eric Nuermberger3, Tjip van der Werf2,4

1  Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine and

I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany

2  Infectious Diseases Unit, Department of Internal Medicine, University of Groningen,

Groningen, The Netherlands

3  Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University,

Baltimore, Maryland, USA

4  Department of Pulmonary Diseases and Tuberculosis, University of Groningen,

(3)
(4)

145 Discussion and Future Perspectives

Ch

ap

te

r 1

1

The treatment of Buruli ulcer (BU), a neglected tropical disease (NTD), has come a long way from primarily surgical care with terrible sequelae, scarring and relapse (1-3) to primarily antimicrobial therapy (4). Current WHO-recommended (5) antimicrobial regimens consist-ing of rifampin (RIF) plus either streptomycin (STR) or clarithromycin (CLR) achieve high cure rates as 8-week regimens (6-8) and patients with small lesions report good quality of life after healing (9). However, lesions vary greatly in size and in form. BU may present as ul-cerative, nodular, oedematous or plaque forms (10). Albeit mostly singular, BU lesions vary greatly in size and are defined by cross-sectional diameter. WHO category I <5cm, category II 5-15cm and category III >15cm, or any lesion on critical locations (e.g. eye, genitalia) or presence of multiple lesions (7). Preliminary results of a study (ClinicalTrials.gov Identifier: NCT01659437) investigating 8 weeks of RIF+CLR showed non-inferiority to the RIF+STR regi-men and spark the hope for soon fully replacing the oto- and nephrotoxic (11) injectable and providing all-oral 8-week therapy.

Despite these advances, further improvement of BU therapy is needed; development of efficient and oral treatment is currently the top BU research priority at WHO, besides unrav-elling the transmission mode and the development of a rapid diagnostic test (5).

Three research items should to be addressed concerning BU therapy: a) the dosing and pharmacokinetics of the current RIF+CLR regimen should be further explored and optimized, b) the treatment duration should be shortened and stratified to lesion category and c) new anti-mycobacterial agents should be evaluated for future shorter-course therapy with quick time to healing of BU lesions.

The current dosage of RIF is based on out-dated arguments about cost-effectiveness and side effects. High-dose RIF is safe in humans (12) and renders BU lesions in mice culture-negative in 4 weeks (13). The widespread availability of the drug and the ease to “double the dose and half the duration” are enticing. Nonetheless, a careful clinical study should evaluate the efficacy in BU patients and pay attention to paradoxical reactions which could arise during rapid killing of bacilli, however were not observed in mice (14). Concerning the macrolide companion, bi-directional drug-drug interactions between RIF and CLR have been noticed (15). Other macrolides such as azithromycin (AZI) (Omansen et al, in prepara-tion) or modern oxazolidinones like sutezolid or tedizolid (16) could be a better option. A recently emerged concept in the related M. tuberculosis infection is that the granulomatous lesion-microenvironment greatly affects local drug penetration and pharmacokinetics (17). Further pre-clinical research is needed to assess the lesion pharmacokinetics of drugs used to treat BU.

(5)

Chapter 11

146

In 2017, despite efforts to promote earlier diagnosis, still 31% of lesions were category III (Chapter 2). However, there are no studies or recommendations on the treatment of large lesions. Albeit it is free of charge in most countries, the long duration of BU treatment causes indirect costs for patients (18) and exposes them to potentially unnecessary side effects. In summary, “a one size fits all” approach for BU antimicrobial therapy should perhaps be reconsidered. Clinical research should evaluate treatment stratified by lesion category, ad-ministering a shorter-course regimen for WHO category I-II e.g. with high dose RIF (13) and either a macrolide companion (AZI, CLR) or another drug such as an oxazolidinone (Almeida et al, in press). Follow-up of patients who defaulted from antimicrobial therapy revealed healing with sub-8-week treatment duration with regular RIF doses, further supporting the case for shorter-course therapy (19). Management of category III lesions in contrast, should be intensified by paying special attention to wound care, physiotherapy to prevent sequelae and psychosocial counselling for stigma prevention. Clofazimine (CFZ) is an off-patent drug that is mycobacteriocidal and has been shown to have good efficacy in experimental M.

ulcerans mouse studies (20). Its availability is expected to increase as a result of its new

place in shorter-course regimen for multidrug-resistant tuberculosis recently recommended by WHO. CFZ could be administered in addition to the abovementioned two drugs. A total duration of 6-8 weeks seems necessary considering the fact that the lesions may sometimes be extensive (Table 1). All abovementioned suggestions can be implemented rather easily and rapidly as all stated drugs are marketed, well-known and available, with the exception of the newer oxazolidinones.

Research for new anti-mycobacterial agents, especially for the treatment of M. tuberculosis infection has produced several compounds that could be applied for the therapy of BU in the future, specifically Q203, bedaquiline (BDQ), its newer analogues 587 and TBAJ-876, and an injectable long-acting formulation of BDQ (LA-BDQ). Q203 is an inhibitor of the cytochrome bc1 complex (specifically, the QcrB component), a 4-gene complex in the mycobacterial electron transport chain responsible for oxidative phosphorylation (21). M.

tuberculosis is able to circumvent this mechanism by changing to a different metabolism,

namely the use of bd oxidase; in M. ulcerans genes encoding for the bd oxidase are non-functional pseudogenes. The MIC of Q203 against M. ulcerans is 0.000078-0.00015 μg/ml (22) and the compound has been shown to be highly bactericidal against M. ulcerans in mice using monotherapy (23); combination regimens e.g. with high-dose rifampin or other drugs prove to be very efficient (24). The rapid microbiological killing of M. ulcerans by Q203 is only surpassed by its rapid reduction of lesion swelling, possibly owing to secondary anti-inflammatory properties of the compound (21,24). BDQ is an inhibitor of the mycobacterial ATP synthase employed in the therapy of tuberculosis (25). BDQ is also highly active against

(6)

147 Discussion and Future Perspectives

Ch

ap

te

r 1

1

M. ulcerans with an MIC of 0.06-0.125 µg/ml (22), comparable to its MIC against M. tuber-culosis. The newer BDQ analogues TBAJ-587 and TBAJ-876 exhibit the 4-8x greater potency

compared to BDQ against M. tuberculosis and are thus of interest for further exploration in BU, too. In an effort to develop short and efficient BU therapy options we are currently testing the use of both oral and LAI-BDQ. LAI-BDQ is a slow-release, i.m. injectable formu-lation. Antibacterial activity could still be observed 12 weeks after a single injection in a mouse model of latent tuberculosis infection (26). Contemplation of shorter-course, highly efficient BU therapy comes with the realization that mycobacterial killing alone does not heal an ulcer instantaneously; culture-negativity is a success from a microbiological view-point but adequate healing without stigmatizing scarring and impairment of locomotion (27) must be the primary endpoint of BU therapy. Perhaps, the use of anti-inflammatory agents as adjuvants can be considered, so lesion healing can be matched by bacterial kill-ing. Although macrolides have been shown to exhibit immunomodulatory effects (28), no differences in sequelae could be detected in an earlier randomized study (7); that study was obviously not designed or powered to detect such immunomodulatory effects. Adjunct anti-inflammatory therapy is considered in M. tuberculosis infection (ClinicalTrials.gov Identifier: NCT03160638). Although some authors have argued in favour of corticosteroids to treat the paradoxical inflammatory response in BU (29), the natural course of these paradoxical responses is usually benign and self-limiting (30).

Lastly, efficient antimicrobial therapy must be accompanied by adequate non-medical treat-ment, such as physiotherapy to prevent sequelae, psychosocial counselling to reduce the burden of stigma, especially for cases with large lesions or lesions in visible critical locations, such as the face, as well as by efficient pain management (31). Integration of the care for BU with other NTDs will offer opportunities for joint engagement of skin NTDs (32,33).

Even though surgery has been shown to be unnecessary in many cases of BU (34), which we hypothesize will be even more so with the advent of new highly active drugs, it should not be neglected but, rather, carefully used in complicated and very advanced cases.

M. ulcerans is an environmental pathogen with an unknown niche and supposedly multiple

transmission pathways (10); the changing epidemiology of BU obeys unknown environmen-tal drivers (35). It is paramount to thus realise that BU will likely never be eradicated, yet it can be controlled well. Fast, reliable and efficient antimicrobial therapy is a key element for efficient disease control in BU and research should focus on improving it. Drug development is a costly process and it is difficult to gain funding for NTD research. Clinical trials for BU are further complicated by the declining global burden of BU and its occurrence in isolated

(7)

Chapter 11

148

foci, even though local epidemics arise such as in Australia (Chapter 2). International col-laboration between researchers and clinicians, coordination and support through WHO, as well as engagement of NGOs and industry partners will be indispensable to translate the promising aforementioned pre-clinical findings into the clinical setting to form the future of Buruli ulcer treatment.

Table 1: Suggested future treatment algorithm for Buruli ulcer; therapy stratified by lesion category with high-dose

rifampin plus either macrolide or oxazolidinone for category I-II and for category III lesions addition of clofazimine. Overview of potential drug candidates worthy of further clinical investigation for Buruli ulcer treatment. RIF, ri-fampin, CFZ, clofazimine, BDQ, Bedaquiline.

lesion category antimicrobial therapy non-medical treatment WhO I – II 2-4 weeks

High-dose RIF + macrolide/ oxazolidinone

Wound care Pain management Physiotherapy as needed

WhO III 8 weeks

High-dose RIF + macrolide/ oxazolidinone + CFZ

Advanced wound care Pain management

Surgery if necessary Intensified Physiotherapy Psychosocial counselling

Future drug candidates

Q203 Cytochrome bc1 inhibitor, MIC 0.000078-0.00015 µg/ml

BdQ and analogues Diarylquinolines, BDQ MIC 0.06-0.125 µg/ml, newer analogues have 4-8x more potent activity

long-acting injectable BdQ One-time i.m. injections offer up to 12 weeks slow release and antibacterial activity

(8)

149 Discussion and Future Perspectives

Ch ap te r 1 1 reFerenCes

1. Stienstra Y, van Roest MHG, van Wezel MJ, Wiersma IC, Hospers IC, Dijkstra PU, et al. Factors associated with functional limitations and subsequent employment or schooling in Buruli ulcer patients. Trop Med Int Health. 2005 Dec; 10(12): 1251–7.

2. Debacker M, Aguiar J, Steunou C, Zinsou C, Meyers WM, Portaels F. Buruli ulcer recurrence, Benin. Emerg-ing Infect Dis. 2005 Apr; 11(4): 584–9.

3. Converse PJ, Nuermberger EL, Almeida DV, Grosset JH. Treating Mycobacterium ulcerans disease (Buruli ulcer): from surgery to antibiotics, is the pill mightier than the knife? Future Microbiol. 2011 Oct; 6(10): 1185–98.

4. Yotsu RR, Richardson M, Ishii N. Drugs for treating Buruli ulcer (Mycobacterium ulcerans disease). Cochrane Infectious Diseases Group, editor. Cochrane Database Syst Rev. 2018 Aug 23; 8(4): CD012118.

5. World Health Organization. Buruli Ulcer (Mycobacterium Ulcerans Infection). 2018 [cited 2018 Dec 13]. Available from: https: //www.who.int/news-room/fact-sheets/detail/buruli-ulcer-(mycobacterium-ulcerans-infection)

6. Etuaful S, Carbonnelle B, Grosset J, Lucas S, Horsfield C, Phillips R, et al. Efficacy of the combination rifampin-streptomycin in preventing growth of Mycobacterium ulcerans in early lesions of Buruli ulcer in humans. Antimicrob Agents Chemother. American Society for Microbiology; 2005 Aug; 49(8): 3182–6. 7. Nienhuis WA, Stienstra Y, Thompson WA, Awuah PC, Abass KM, Tuah W, et al. Antimicrobial treatment

for early, limited Mycobacterium ulcerans infection: a randomised controlled trial. Lancet. 2010 Feb 20; 375(9715): 664–72.

8. Phillips RO, Sarfo FS, Abass MK, Abotsi J, Wilson T, Forson M, et al. Clinical and bacteriological efficacy of rifampin-streptomycin combination for two weeks followed by rifampin and clarithromycin for six weeks for treatment of Mycobacterium ulcerans disease. Antimicrob Agents Chemother. 2014; 58(2): 1161–6. 9. Klis S, Ranchor A, Phillips RO, Abass KM, Tuah W, Loth S, et al. Good quality of life in former Buruli ulcer

patients with small lesions: long-term follow-up of the BURULICO trial. Johnson C, editor. PLoS Negl Trop Dis. 2014 Jul; 8(7): e2964.

10. Yotsu RR, Suzuki K, Simmonds RE, Bedimo R, Ablordey A, Yeboah-Manu D, et al. Buruli Ulcer: a Review of the Current Knowledge. Curr Trop Med Rep. 2018; 5(4): 247–56.

11. Klis S, Stienstra Y, Phillips RO, Abass KM, Tuah W, van der Werf TS. Long term streptomycin toxicity in the treatment of Buruli Ulcer: follow-up of participants in the BURULICO drug trial. Johnson C, editor. PLoS Negl Trop Dis. 2014 Mar; 8(3): e2739.

12. Boeree MJ, Diacon AH, Dawson R, Narunsky K, Bois du J, Venter A, et al. A Dose-Ranging Trial to Optimize the Dose of Rifampin in the Treatment of Tuberculosis. Am J Respir Crit Care Med. 2015 May; 191(9): 1058–65.

13. Omansen TF, Almeida D, Converse PJ, Li S-Y, Lee J, Stienstra Y, et al. High-dose rifamycins enable shorter oral treatment in a murine model of Mycobacterium ulcerans disease. 2018 Jan 1.

14. Omansen TF, Almeida D, Converse PJ, Li S-Y, Lee J, Stienstra Y, et al. High-dose rifamycins enable shorter oral treatment in a murine model of Mycobacterium ulcerans disease. Antimicrob Agents Chemother. 2018 Nov 19.

15. Alffenaar JWC, Nienhuis WA, de Velde F, Zuur AT, Wessels AMA, Almeida D, et al. Pharmacokinetics of Rifampin and Clarithromycin in Patients Treated for Mycobacterium ulcerans Infection. Antimicrob Agents Chemother. American Society for Microbiology; 2010 Sep 1; 54(9): 3878–83.

(9)

Chapter 11

150

16. Almeida DV, Omansen TF, Li S-Y, Lee J, Grosset JH, Converse PJ, et al. Oxazolidinones Can Replace Clarithro-mycin in Combination with Rifampin in a Mouse Model of Buruli Ulcer. Antimicrob Agents Chemother. 2018 Dec 17.

17. Prideaux B, Via LE, Zimmerman MD, Eum S, Sarathy J, O’Brien P, et al. The association between sterilizing activity and drug distribution into tuberculosis lesions. Nat Med. 2015 Oct; 21(10): 1223–7.

18. Amoakoh HB, Aikins M. Household cost of out-patient treatment of Buruli ulcer in Ghana: a case study of Obom in Ga South Municipality. BMC Health Serv Res. 2nd ed. BioMed Central; 2013 Dec 5; 13(1): 507. 19. Klis S, Kingma RA, Tuah W, van der Werf TS, Stienstra Y. Clinical outcomes of Ghanaian Buruli ulcer patients

who defaulted from antimicrobial therapy. Trop Med Int Health. 2016 Sep; 21(9): 1191–6.

20. Converse PJ, Almeida DV, Tasneen R, Saini V, Tyagi S, Ammerman NC, et al. Shorter-course treatment for Mycobacterium ulcerans disease with high-dose rifamycins and clofazimine in a mouse model of Buruli ulcer. Small PLC, editor. PLoS Negl Trop Dis. 2018 Aug 13; 12(8): e0006728.

21. Pethe K, Bifani P, Jang J, Kang S, Park S, Ahn S, et al. Discovery of Q203, a potent clinical candidate for the treatment of tuberculosis. Nat Med. 2013 Sep; 19(9): 1157–60.

22. Almeida DV. Johns Hopkins University. Center For Tuberculosis Research. November 2018. Personal com-munication.

23. Scherr N, Bieri R, Thomas SS, Chauffour A, Kalia NP, Schneide P, et al. Targeting the Mycobacterium ulcerans cytochrome bc1: aa3 for the treatment of Buruli ulcer. Nat Commun. 2018 Dec 18; 9(1): 5370.

24. Converse P. Johns Hopkins University. Center For Tuberculosis Research. November 2018. Personal com-munication.

25. Diacon AH, Pym A, Grobusch MP, de los Rios JM, Gotuzzo E, Vasilyeva I, et al. Multidrug-resistant tubercu-losis and culture conversion with bedaquiline. N Engl J Med. 2014 Aug 21; 371(8): 723–32.

26. Kaushik A. Johns Hopkins University. Center For Tuberculosis Research. November 2018. Personal com-munication.

27. de Zeeuw J, Omansen TF, Douwstra M, Barogui YT, Agossadou C, Sopoh GE, et al. Persisting social participa-tion restricparticipa-tions among former Buruli ulcer patients in Ghana and Benin. Small PLC, editor. PLoS Negl Trop Dis. Public Library of Science; 2014 Nov; 8(11): e3303.

28. Altenburg J, de Graaff CS, van der Werf TS, Boersma WG. Immunomodulatory effects of macrolide antibiot-ics - part 1: biological mechanisms. Respiration. 2011; 81(1): 67–74.

29. Trevillyan JM, Johnson PDR. Steroids control paradoxical worsening of Mycobacterium ulcerans infection following initiation of antibiotic therapy. The Medical Journal of Australia. 2013 May 6; 198(8): 443–4. 30. Nienhuis WA, Stienstra Y, Abass KM, Tuah W, Thompson WA, Awuah PC, et al. Paradoxical responses after

start of antimicrobial treatment in Mycobacterium ulcerans infection. Clin Infect Dis. 2012 Feb 15; 54(4): 519–26.

31. Alferink M, de Zeeuw J, Sopoh G, Agossadou C, Abass KM, Phillips RO, et al. Pain Associated with Wound Care Treatment among Buruli Ulcer Patients from Ghana and Benin. Hausmann-Muela SC, editor. PLoS ONE. 2015; 10(6): e0119926.

32. Barogui YT, Diez G, Anagonou E, Johnson RC, Gomido IC, Amoukpo H, et al. Integrated approach in the control and management of skin neglected tropical diseases in Lalo, Benin. Small PLC, editor. PLoS Negl Trop Dis. 2018 Jun; 12(6): e0006584.

33. Engelman D, Fuller LC, Solomon AW, McCarthy JS, Hay RJ, Lammie PJ, et al. Opportunities for Integrated Control of Neglected Tropical Diseases That Affect the Skin. Trends Parasitol. 2016 Nov; 32(11): 843–54.

(10)

151 Discussion and Future Perspectives

Ch

ap

te

r 1

1

34. Wadagni AC, Barogui YT, Johnson RC, Sopoh GE, Affolabi D, van der Werf TS, et al. Delayed versus standard assessment for excision surgery in patients with Buruli ulcer in Benin: a randomised controlled trial. Lancet Infect Dis. 2018 Jun; 18(6): 650–6.

35. O’Brien DP, Jeanne I, Blasdell K, Avumegah M, Athan E. The changing epidemiology worldwide of Mycobac-terium ulcerans. Epidemiol Infect. 2018 Oct 8; 187: 1–8.

(11)

Referenties

GERELATEERDE DOCUMENTEN

The delayed healing following antimicrobial therapy of Buruli ulcer is believed to have led to an underappreciation of the value of antimicrobial in Buruli ulcer management in

Cotrimoxazol in the treatment of Mycobacterium ulcerans infection (Buruli ulcer) in west Africa. Espey DK, Djomand G, Diomande I, Dosso M, Saki MZ, Kanga J-M, et al. A pilot study

In this pilot study, we developed this idea further and tested IVIS® imaging of experimentally infected mice with the aim to refine the mouse tail model of Buruli ulcer and to

Following on from reports that avermectins have activity against Mycobacterium tuberculosis, we tested the in-vitro efficacy of ivermectin and moxidectin on M.. We observed

Accumulation observed as increasing trough concentrations after several days of IVM administration (FIG 1C) as well as the previously reported lipophilicity and accumulation

Figure 1: Median of footpad swelling grade of infected mouse footpads in response to treatment with high-dose rifamycins and clarithromycin.. Treatment was initiated 6 weeks

In this experiment, we compared the efficacy of these oxazolidinones, including a reduced dose of LZD, in combination with RIF to that of standard-of-care regimens based on their

ulcerans BALB/c mouse model that yielded high- dose rifampin as high-potential candidate regimen for further evaluation of future highly active, short-course regimen to treat BU,