Next Article in Journal
The Multi-Strain Probiotic OMNi-BiOTiC® Active Reduces the Duration of Acute Upper Respiratory Disease in Older People: A Double-Blind, Randomised, Controlled Clinical Trial
Next Article in Special Issue
Tick Species Diversity and Molecular Identification of Spotted Fever Group Rickettsiae Collected from Migratory Birds Arriving from Africa
Previous Article in Journal
Kosakonia cowanii Ch1 Isolated from Mexican Chili Powder Reveals Growth Inhibition of Phytopathogenic Fungi
Previous Article in Special Issue
Molecular Evidence of Rickettsia conorii subsp. raoultii and Rickettsia felis in Haemaphysalis intermedia Ticks in Sirumalai, Eastern Ghats, Tamil Nadu, South India
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Use of Natural Bioactive Nutraceuticals in the Management of Tick-Borne Illnesses

by
Samuel M. Shor
1,* and
Sunjya K. Schweig
2
1
Internal Medicine of Northern Virginia, George Washington University Health Care Sciences, Reston, VA 20190, USA
2
California Center for Functional Medicine, Oakland, CA 94619, USA
*
Author to whom correspondence should be addressed.
Microorganisms 2023, 11(7), 1759; https://doi.org/10.3390/microorganisms11071759
Submission received: 14 May 2023 / Revised: 29 June 2023 / Accepted: 29 June 2023 / Published: 5 July 2023
(This article belongs to the Special Issue Emerging Research on Tick-Borne Pathogens and Diseases)

Abstract

:
The primary objective of this paper is to provide an evidence-based update of the literature on the use of bioactive phytochemicals, nutraceuticals, and micronutrients (dietary supplements that provide health benefits beyond their nutritional value) in the management of persistent cases of Borrelia burgdorferi infection (Lyme disease) and two other tick-borne pathogens, Babesia and Bartonella species. Recent studies have advanced our understanding of the pathophysiology and mechanisms of persistent infections. These advances have increasingly enabled clinicians and patients to utilize a wider set of options to manage these frequently disabling conditions. This broader toolkit holds the promise of simultaneously improving treatment outcomes and helping to decrease our reliance on the long-term use of pharmaceutical antimicrobials and antibiotics in the treatment of tick-borne pathogens such as Borrelia burgdorferi, Babesia, and Bartonella.

1. Introduction

Lyme disease results from an active infection from any of several pathogenic members of the Borrelia burgdorferi sensu lato complex (Bbsl), including Borrelia burgdorferi sensu stricto (Bbss) and Borrelia mayonii (in North America) and Borrelia burgdorferi sensu strictu, Borrelia afzelii, and Borrelia garinii (in Eurasia). Lyme disease is the most common vector-borne illness in the United States [1] and Europe [2], and the Centers for Disease Control and Prevention (CDC) estimates that the annual incidence of Lyme disease in the US is at least 476,000 [3,4].
While a standard course of antibiotics may effectively treat many people with acute Lyme disease, there is mounting evidence that a typical course of antibiotics for acute Lyme disease may leave up to 15–35% of people with chronic symptoms and health issues [5]. While the exact reasons for this ongoing illness remain an area of active research and discussion, various investigators point to persistent infection as a possible and important cause. In categorizing chronic active Lyme disease (CLD) cases, a consensus publication described two categories. The first category, CLD-U, describes an untreated group with clinical features consistent with Lyme disease, likely representing active infection, that for various reasons was never identified and/or treated. The second category, CLD-PT, or the “previously treated” group, represents those appropriately identified, but for which the treatment was inadequate. In this group, suspected active infection was thought to explain ongoing clinical features. Both groups were symptomatic for at least 6 months [6].
The pathophysiology of chronic active (CLD-PT) Bburgdorferi infection [6] is supported by both animal and human persistence studies [7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33]. Studies have shown that B. burgdorferi species can exist in three morphological forms. The typical motile corkscrew spirochete represents the vegetative (active) form. Under stress conditions, including changes in pH, osmolarity, and antibiotic exposure in vitro, the motile spirochetes can transform into latent persistent stationary forms and/or biofilm-like aggregates [34,35,36,37,38,39,40,41]. Additional survival mechanisms of B. burgdorferi persistence include “immune evasion via physical seclusion within immunologically protected tissue sites such as the CNS, joints and eyes, collagen-rich tissues, cells and biofilms; alterations in outer surface protein (OSP) profiles through antigenic variation, phasic variations, immune modulation via alterations in complement, neutrophil and dendritic cell functioning, and changes in cytokine and chemokine levels” [6].
Biofilm generation by pathogenic bacteria, including B. burgdorferi species [36,42], is another well-known mechanism of microbe persistence which can potentially lead to CLD. The presence of B. afzelii embedded in biofilm has been demonstrated in infected human tissues [42]. Of those diagnosed with tick-borne diseases, growing numbers are being found to have polymicrobial infections [43,44,45,46]. One such pathogen is the malaria-like intraerythrocytic parasite Babesia [47,48,49,50]. Research by Dunn and colleagues [48] suggests that the concomitant presence of B. burgdorferi in ticks contributes to the emergence and geographic expansion of Babesia microti. Further, those infected with multiple tick-borne pathogens tend to have more severe courses [51], contributing to increased challenges in achieving recovery.
The cornerstone of therapy for tick-borne infections has been antimicrobials. The value of using prolonged antibiotic courses is controversial [52,53,54], with differing recommendations. Guidelines and clinicians recommending shorter durations of treatment cite a perceived lack of therapeutic benefit. This impression is largely based on their interpretation of the findings of four NIH-funded trials [55,56,57]. However, the first two studies [55] were felt to have significant design and methodology flaws, resulting in questions as to their generalizability [58,59]. Importantly, the other two studies had sub-cohort analysis supporting the potential value of longer duration antimicrobial therapy [56,57].
Potential B. burgdorferi persistence causing chronic infection affirms the need for longer duration treatment than the current IDSA recommendations [54,58,60]. In our opinion, the clinical course and required treatment duration are dependent upon many factors. These include the severity of the individual case, the duration of illness prior to treatment, comorbidities, and the presence of coinfections or opportunistic infections [43,44]. Rather than using an arbitrary, “one size fits all” guideline [52,53], analysis and treatment decisions are made by the clinician at the point of care, using their clinical expertise and judgement [54].
Precedence exists for the appropriateness and necessity of prolonged courses of antimicrobials in clinical practice. Examples include multidrug-resistant tuberculosis, where the recommended treatment duration can be up to 15–24 months after culture conversion [61]. Additional examples include histoid leprosy, caused by Mycobacterium leprae, which requires 12–24 months of therapy [62], and Q fever endocarditis caused by the organism Coxiella burnetti, where one study showed that most patients required 18–24 months of treatment to sterilize their infected heart valves [63].
Due to the frequent polymicrobial and persistent nature of many tick-borne infections, treatment often relies upon prolonged duration and multidrug protocols [64]. However, it is critical to balance optimal antibiotic stewardship and the goal of minimizing the overuse of antibiotics, with the potential risk of driving antibiotic resistance. By understanding the pathophysiology of persistent Lyme disease and other tick-borne illnesses and using botanical medicines and nutraceuticals to target pleomorphic persister cells and biofilms [33,34,35,36,37,38,39,40,41,42], we have the potential to be more judicious in the use of prescription antibiotics. In so doing, we also have the greater potential to decrease the duration and complexity of antibiotic protocols, while improving outcomes. In addition, botanical medicines and nutraceuticals can have antimicrobial efficacy and potency against persister forms and thus have the potential to treat tick-borne illnesses independently of prescription antibiotics.

2. Methods

Using PUB MED, comprehensive searches were employed including using the following search terms, “Lyme disease”, “Borrelia”, “Bartonella”, “Babesia”, and cross referencing with generic concepts such as “herbals”, “nutraceuticals”, and “botanical medicines” along with each of the multiple agents described in this manuscript. Over 400 references were identified and reviewed. To meet inclusion criteria, studies had to include agents shown to have a therapeutic effect on at least one of the following: Borrelia burgdorferi and/or Bartonella species in active, stationary or biofilm forms, or activity against Babesia strains. Papers not demonstrating treatment evidence against any of these pathogens were excluded and thus not referenced.

3. Discussion

Botanical medicines and natural antimicrobial agents have been used for thousands of years and have been shown to be effective against various pathogens [65]. Goc and colleagues [66] tested 15 phytochemicals and micronutrients for activity against three morphological forms of Borrelia burgdorferi and Borrelia garinii (spirochetes, latent rounded forms, and biofilm). Their results showed that the most potent substances against the spirochete and rounded forms of B. burgdorferi and B. garinii were cis-2-decenoic acid, baicalein, monolaurin, and kelp (iodine), and that only baicalein and monolaurin revealed significant activity against the biofilm form [66]. Table 1 reflects a summary of organic oils studied by Goc and colleagues [67].
Feng and colleagues [68] investigated numerous agents for potential in vitro anti-Borrelia burgdorferi activity. Among these natural products, seven were found to have good activity against the stationary phase B. burgdorferi culture compared to the control antibiotics doxycycline and cefuroxime. The identified botanicals included Cryptolepis sanguinolenta, Juglans nigra (Black walnut), Polygonum cuspidatum (Japanese knotweed), Artemisia annua (Sweet wormwood), Uncaria tomentosa (Cat’s claw), Cistus incanus, and Scutellaria baicalensis (Chinese skullcap) [68].
Some essential oils extracted from plants have also been reported in the literature to have antimicrobial activities [69,70,71,72]. In a more recent in vitro essential oil study, Feng and colleagues [73] evaluated the activity of 34 essential oils in a B. burgdorferi stationary phase culture persister model. In this study, the top five essential oils (oregano, cinnamon bark, clove bud, citronella, and wintergreen) showed high anti-persister activity. Importantly, some of the essential oils were found to have excellent anti-biofilm ability, as shown by their ability to dissolve the aggregated biofilm-like structures. The top three hits, oregano, cinnamon bark, and clove bud, completely eradicated all viable cells without any regrowth in subcultures in fresh medium.
Furthermore, a number of researchers have explored the utility of botanical medicines for the treatment of Babesia infections [74,75,76,77]. According to Zhang e and colleague [78], some botanical medicines and their active constituents have potent activity against B. duncani in vitro and may be further explored for the more effective treatment of babesiosis. In a recent study that used a hamster erythrocyte model, a panel of herbal medicines was screened and Cryptolepis sanguinolenta, Artemisia annua, Scutellaria baicalensis, Alchornea cordifolia, and Polygonum cuspidatum were identified to have good in vitro inhibitory activity against B. duncani [74]. In another study, Zhang and colleagues [77] described how garlic oil and black pepper oil were active against B. duncani. Furthermore, Batiha and colleagues [76] found that C. verum extracts were potential anti-piroplasm drugs through in vitro and animal in vivo models.
Bartonella infection had previously been thought to only be responsible for an acute, self-limiting illness. One such example is cat-scratch disease. Caused by Bartonella henselae transmitted during a cat bite or scratch, this can frequently manifest as an acute self-limited febrile illness associated with regional lymphadenopathy. However, evidence is now supporting a more complex perspective where subsets of infected, immunocompetent patients can become chronically bacteremic [78,79,80]. There is no single treatment effective for systemic B. henselae infection, and antibiotic therapy exhibited poor activity against typical uncomplicated cat scratch disease [81]. With possible persistent infection, Bartonella spp. infections can represent a significant treatment challenge [81]. Similar to Borrelia, stationary phase and biofilm persister cells have been identified with Bartonella henselae [82,83,84,85]. Standard treatments have a relatively poor impact on persister cells, and this is likely one important factor contributing to treatment failure and the persistence of infection [82].
Creative approaches to address mechanisms of persistence have been studied. Li and colleagues [84] worked to identify FDA-approved drugs with activity against stationary phase Bartonella henselae. In their study, they were able to show that pyrantel pamoate, daptomycin, methylene blue, clotrimazole, gentamicin, and streptomycin, at their respective maximum serum drug concentration (Cmax), had the capacity to completely eradicate stationary phase B. henselae after 3 days’ drug exposure in subculture studies. In this study, while the currently used drugs for treating bartonellosis, including rifampin, erythromycin, azithromycin, doxycycline, and ciprofloxacin, had very low minimal inhibitory concentration (MIC) against growing B. henselae, they had relatively poor activity against stationary phase B. henselae [84]. Given these findings, there has been growing interest in identifying additional active agents to treat Bartonella infections and the use of phytochemicals has been increasingly explored [82,84,85]. For example, two active ingredients of the essential oils oregano and cinnamon bark, carvacrol and cinnamaldehyde, respectively, were shown to be highly active against the stationary phase B. henselae and able to eradicate all the bacterial cells [82].
Appendix A provides an overview of many of the nutraceuticals studied. Included in this summary are agents identified as the most effective agents in a particular study. The key to these data with appropriate references can be reviewed in Appendix B.

3.1. Specific Agents

3.1.1. Alchornea cordifolia Extracts

Alchornea cordifolia has documented antimicrobial and anti-inflammatory activity [86,87]. In addition, A. cordifolia has significant antimalarial effects [88,89,90] and has been used by traditional herbalists for the treatment of malaria in several African countries [91]. According to Zhang and colleagues [74], this botanical medicine showed good inhibitory effects against Babesia duncani. Importantly, preclinical studies revealed that Alchornea cordifolia has favorable toxicology and bioavailability profiles [91,92,93,94].

3.1.2. Allicin (Garlic)

Allicin represents a key component of garlic, with a broad spectrum of antimicrobial activities [95]. These actions include (i) a therapeutic effect against a wide range of Gram-negative and Gram-positive bacteria; (ii) antifungal activity, such as Candida albicans; (iii) antiparasitic activity, such as Entamoeba histolytica and Giardia lamblia; and (iv) antiviral activity [95]. According to Feng, when assessing the activity of 35 essential oils in eradicating B. burgdorferi stationary forms in vitro, “garlic essential oil exhibited the best activity as shown by the lowest residual viability of B. burgdorferi [68,96]”. Zhang and colleagues [77] further described that “garlic oil and black pepper oil showed the highest activity against B. duncani growth”. According to Salama and colleagues [97], in an in vivo murine model, parasitemia of Babesia microti significantly decreased in allicin-treated mice (p < 0.01) from days 4–14 post-inoculation, as compared to controls.

3.1.3. Andrographis paniculata

While a recent in vitro study by Feng and colleagues [68] did not support the efficacy of Andrographis paniculata against Borrelia species, it does have previous research to support its efficacy against Leptospirosis, another spirochetal illness [98].

3.1.4. Artemisia annua

Also called sweet wormwood, Chinese wormwood, and Qing Hao, Artemesis annua has been used for over 2000 years [99] with particular attention to treating malaria [100]. Active components of Artemesia annua include artemisinin [101], artesunate, and artemether, all of which are potent antimalarial drugs [102]. The active ingredient artemisinin has been shown to have in vitro activity against stationary phase B. burgdorferi persister cells [103,104]. In humans, Artemisia annua has been used safely in doses up to 2250 mg daily for up to 10 weeks, and 1800 mg daily has also been used safely for up to six months [105]. At higher doses, gastrointestinal upset has been reported, including mild nausea, vomiting (rarer), and abdominal pain.

3.1.5. Berberis vulgaris/Berberine

Berberis vulgaris (B. vulgaris), is naturally found in semi-tropical areas in Africa, Asia, Europe, North America, and South America. Pharmacologically, B. vulgaris has been shown to have numerous properties, including anti-inflammatory, sedative, antipyretic, antiemetic, antioxidant, anti-cholinergic, anti-arrhythmic, and antimicrobial (including anti-malarial) properties [106,107]. Two active ingredients, berberine and berbamine, further demonstrate hypoglycemic, anti-inflammatory, hypotensive, antioxidant, and hypolipidemic properties [108,109]. Antimicrobial effects have been reported against multiple pathogens, including Trichomonas vaginalis, Giardia lamblia, Entamoeba histolytica, and certain Leishmania strains. According to Subecki and colleagues [110], berberine was shown to have inhibitory effects against Babesia gibsoni. According to Batiha and colleagues [111], a methanolic extract of B. vulgaris (MEBV) restricted the multiplication of several Babesia species, including B. bovis, B. bigemina, B. divergens, and B. cabali. Elkhateeb and colleagues [112] reviewed the anti-babesial activity of nine North African medicinal plants against the growth of Babesia gibsoni in vitro. Extracts prepared from Berberis vulgaris and Rosa damascene showed more than 90% inhibition at a concentration of 100 mcg/mL. Multiple isolates were shown to be active against B. vulgaris, with the most potent being syringic acid. Finally, berberine has been shown by Li and colleagues [84] to have relatively high activity against stationary phase Bartonella henselae.

3.1.6. Cinnamomum (Cinnamon)

Cinnamon is a tropical Asian spice and a native plant of Sri Lanka and is extracted from the Cinnamomum genus, including Cinnamomum camphora, Cinnamomum osmophloeum, Cinnamomum burmannii, Cinnamomum zeylanicum, Cinnamomum cassia, and Cinnamomum verum [113]. Cinnamaldehyde, a key active ingredient, has been shown to have efficacy against the active spirochetal [67,96] and stationary forms of Borrelia burgdorferi sensu latu [67,73,96], as well as to the bio-film forms [67]. Feng and colleagues [73] found that essential oil from cinnamon bark (at a low concentration of 0.25%) was one of five essential oils with high anti-persister activity and better activity than the known persister drug daptomycin. Further, Feng and colleagues [73] reported that cinnamon bark had anti-Borrelia biofilm activity.
With regard to non-Borrelial tick borne pathogens, Ma and colleagues [82] reported that cinnamon bark and cinnamon leaf were both active against the stationary phase non-growing B. henselae, and also had good activity against log-phase growing B. henselae. Batiha and colleagues [76] provided evidence that several Babesia strains were inhibited in vitro by derivatives of cinnamon including B. bovis, B. bigemina, B. divergens, B. caballi, and T. equi. Furthermore, this study also showed that cinnamon was effective in vivo against Babesia microti.

3.1.7. Cistus creticus

In vitro studies have shown that Cistus species have antibacterial, antifungal, antiviral, and anti-inflammatory properties [114,115,116,117,118,119,120,121,122]. In one study, volatile oil from Cistus creticus was shown to inhibit growing forms of B. burgdorferi. This study also found that essential oil volatile compounds exhibited a stronger growth inhibitory effect than leaf extracts [123]. Feng and colleagues [68,73] reported that Carvacrol was likely the active Cistus creticus component against log and stationary phase Borrelia burgdorferi.

3.1.8. Cryptolepis sanguinolenta

Pharmacologic properties of Cryptolepis sanguinolenta have been shown to include anti-inflammatory, antimicrobial, anti-amoebic, anti-cancer, and anti-malarial features [124,125,126,127,128]. Cryptolepine, an active ingredient of Cryptolepis sanguinolenta, has been shown in vitro to have significant anti-plasmodial activity against drug-sensitive and drug resistant malaria strains [129,130].
Further, two open label trials using different formulations of Cryptolepis sanguinolenta have provided both efficacy and safety in the setting of treating patients with uncomplicated malaria [131,132]. Zhang and colleagues [74] provided in vitro evidence that C. sanguinolenta had efficacy against Borrelia burgdorferi, Bartonella henselae, and Babesia duncani, with good efficacy against both growing and stationary forms. Ma and colleagues [133] also identified efficacy against Bartonella stationary and active log phase forms. Of multiple agents studied, cryptolepine had the strongest inhibitory activity against Babesia duncani, including traditional combinations of quinine and clindamycin. Cryptolepis sanguinolenta has been shown to be safe at doses under 500 mg/kg of body weight [129]. However, the question regarding potential anti-fertility and embryotoxic effects [134,135] needs further study.

3.1.9. Dipsacus sylvestris/Dipsacus fullonum (Teasel Root)

Dipsacus is a species of plant native to Eurasia and North Africa, otherwise known as wild teasel or fuller’s teasel. Leopold and colleagues [136] studied several preparations of this agent against Borrelia burgdorferi sensu strictu. The hydroethanolic extract showed no growth inhibition, whereas significant growth-inhibiting activity could be shown in the two less polar fractions. Liebold and colleagues [136] did not study the efficacy of teasel root extract against round body forms or biofilm. However, Goc and Roth [137] did, finding no impact on persister forms of Borrelia.

3.1.10. Eugenia caryophyllata (Syzigium aromaticum L. (Myrtaceae)

Eugenia caryophyllata, also known as clove, is a dried flower bud belonging to the Myrtaceae family that is indigenous to the Maluku islands in Indonesia [138]. Several important active components have been isolated in clover oil, including eugenyl acetate, eugenol, and β-caryophyllene. Many studies have examined S. aromaticum activity against various pathogenic parasites and microorganisms, including Plasmodium, Babesia, Theileria parasites, Herpes simplex, and hepatitis C viruses [138]. Feng and colleagues [73] identified that clove represented one of the top five essential oils studied (oregano, cinnamon bark, clove bud, citronella, and wintergreen) effective at low concentrations against the B. burgdorferi persister cell stationary phase. In fact, these five agents had greater activity than the known anti-persister drug daptomycin. Further, these researchers identified that clove, and cinnamon bark and oregano, were the top three essential oils with excellent Borrelia anti-biofilm effects. In addition, Ma, and colleagues [82] identified clove bud as one of 32 essential oils that had high activity against both the stationary non-growing phase and the log-phase growing forms of Bartonella henselae. Finally, in an in vivo murine model, Batiha and colleagues [139] published a study supporting the clove extract inhibition of Babesia microti.

3.1.11. Grapefruit Seed Extract (GSE)

Goc and Rath [137] showed that GSE had in vitro activity against both stationary and active forms of B. burgdorferi. However, these positive findings contrasted with negative findings by Feng and colleagues [68], who opined that this disparity could have been explained by different GSE formulations, toxins present in the GSE formulations themselves, and/or different Borrelia strains tested.

3.1.12. Juglans nigra (Black Walnut)

There is evidence that Juglans nigra (black walnut) and its constituents have broad therapeutic properties, including antioxidant, antibacterial, antitumor and chemoprotective effects [140,141]. Regarding potential impacts on Borrelia burgdorferi and Borrelia garinii, Juglans nigra has exhibited bacteriostatic activity against log phase spirochetes of B. burgdorferi and B. garinii and bactericidal activity against Borrelia round bodies [137]. Ma and colleagues [133] also identified efficacy against stationary and active log phase forms of Bartonella. Although reports of gastrointestinal disturbance [142] and changes in skin pigmentation have been published [143,144], Juglans nigra is generally well-tolerated and side effects are uncommon. Reports in humans suggest that there may be some allergic cross reactivity in those allergic to tree nuts or walnuts, as well as cases of dermatitis [145].

3.1.13. Monolaurin

Monolaurin first became available as a nutritional formulation in the mid-1960s and today is sold worldwide as a dietary supplement, commonly known by one of its chemical names, glycerol monolaurate (GML). The richest dietary source is coconut oil [146]. Monolaurin has been shown to have antibacterial activity against several Gram-positive strains such as Staphylococcus sp., Corynebacterium sp., Bacillus sp., Listeria sp., and Streptococcus sp. [147]. Goc and colleagues [66] found that baicalein and monolaurin were effective against biofilm-like colonies of both B. burgdorferi and B. garinii.

3.1.14. Nigella sativa (Black Cumin)

Nigella sativa is known for its antioxidant, anti-inflammatory, antibacterial, antiviral, antiparasitic, anticarcinogenic, antiallergic, antidysenteric, and antiulcer effects. The main component within N. sativa is thymoquinone, which has been proven in vitro and in vivo against the growth of the malarial agent Plasmodium berghei in mice. El-Sayed revealed in vivo the efficacy of thymoquinone against Babesia microti in a murine model [148].

3.1.15. Oregano

The herb oregano comes from the dried leaves of Origanum sp. (Mediterranean variety) or Lippia sp. (Mexican variety) [149]. Oregano is widely used in foods and has been designated GRAS (generally recognized as safe) by the FDA. In a small study, 200 mg/day emulsified O. vulgare oil was administered for 6 weeks. Carvacrol has been shown to be the most active component of oregano oil, and in one study showed excellent activity against B. burgdorferi stationary phase cells. This same study showed that other oregano oil compounds, p-cymene and α-terpinene, had no apparent activity. In addition, Feng and colleagues [73] found that oregano was efficacious against Borrelia biofilm microcolonies [73], and that oregano was one of the top five essential oils tested for activity against B. burgdorferi stationary phase culture. Further, these researchers found that carvacrol had efficacy against log phase Borrelia burgdorferi [73]. Ma and colleagues [82] reported that oregano was active against the stationary phase of non-growing B. henselae and had good activity against log-phase growing B. hensela. Of note, oregano has been found to have good blood–brain barrier penetration [150].

3.1.16. Otoba parvifolia (Banderol)

According to Goc and Rath, Otoba parvifolia has been shown to have in vitro efficacy on active and dormant forms of Borrelia burgdorferi sensu stricto. Further, particularly in combination with Uncaria tomentosa, these agents have provided significant effects on all morphological forms of B. burgdorferi [137].

3.1.17. Piper nigrum (Black Pepper)

As previously mentioned, according to Zhang and colleagues [77], “garlic oil and black pepper oil showed the highest activity against B. duncani growth”.

3.1.18. Polygonum cuspidatum (Japanese Knotweed)

Resveratrol represents one of the main active ingredients in P. cuspidatum. This agent has documented anti-tumor, antimicrobial, anti-biofilm, anti-inflammatory, neuroprotective, and cardioprotective effects [151,152,153,154,155,156]. Further, P. cuspidatum showed strong activity against both growing and non-growing stationary phases of B. burgdorferi [68]. According to Zhang and colleagues [74], P. cuspidatum showed a good inhibitory effect against Babesia duncani and Bartonella henselae. Ma and colleagues [133] also identified P. cuspidatum efficacy against Bartonella stationary and active log phase forms [133]. P. cuspidatum has been found to have minimal toxicity in animal and human studies. Gastrointestinal upset and diarrhea can occur, but generally resolve after decreasing or stopping the intake [157].

3.1.19. Rhus coriaria L. (Sumac)

Rhus coriaria L. (Anacardiaceae) is commonly known as sumac. The most conventional use is as a spice, condiment, and flavoring agent, especially in the Mediterranean region. Accumulating evidence supports the antibacterial, antinociceptive, antidiabetic, cardioprotective, neuroprotective, and anticancer effects of this plant. Toxicity studies show that sumac is generally very safe to consume by humans, with little toxicity [158]. Batiha and colleagues [111] studied an acetone extract of R. coriaria (AERC), which was shown to have an inhibitory effect on all Babesia strains tested; AERC suppressed B. bovis, B. bigemina, B. divergens, and B. caballi.

3.1.20. Rosmarinic Acid

Rosmarinic acid is an active component of the Lamiaceae (Labiatae) family and has been associated with anti-inflammatory and antioxidant activity [159]. According to Goc and colleagues [160], the addition of rosmarinic acid in conjunction with either luteolin or baicalein had additive bacteriostatic effects on B. burgdorferi anti-spirochetal activity.

3.1.21. Scuttelaria spp., Baicalin, and Baicalein

Extracted from the roots of Scutellaria baicalensis and Scutellaria lateriflora, the active flavonoid baicalin is a flavone glycoside obtained via the binding of glucuronic acid.
Another flavonoid, baicalin, has been shown to have anti-biofilm properties in other infections [161] and can work synergistically with other antimicrobials [162,163,164]. Specifically relating to tick-borne infections, Goc and colleagues [66] provided in vitro evidence that baicalein exhibited activity against the three known morphologic forms of B. burgdorferi and B. garinii. These forms included log phase spirochetes, latent round bodies, and biofilm. Feng and colleagues [68] independently provided evidence that this agent was effective in inhibiting both the active and stationary forms of Borrelia burgdorferi. In their 2021 paper, Zhang and colleagues [74] provided evidence that baicalein showed good in vitro activity against B. duncani, superior to both quinine and clindamycin. Interestingly, Goc and colleagues [160] have been able to show synergistic outcomes when baicalein was combined with other nutraceuticals, such as luteolin. This combination showed synergistic cooperation in killing knob-/round-shaped persistent forms and additive effects in the eradication of biofilms formed by the Borrelia burgdorferi and B. garinii studied. This combination was able to eliminate ~90% of active and persistent forms, as well as eradicating 50% of the mature Borrelia biofilms. Furthermore, this synergism was able to extend to formal antimicrobials, such as doxycycline. This combination was synergistic in treating all forms of Borrelia, including active spirochetal, stationary, and biofilm persisters.
Scutellaria baicalensis has documented clinical safety [165,166] and baicalein even exhibits a hepato-protective effect in preventing acetaminophen-induced liver injury [167]. There have been reports of sedation associated with activity on GABA receptor sites [168].

3.1.22. Stevia rebaudiana

Certain formulations of stevia have been shown to be effective against the active, stationary, and biofilm forms of B. burgdorferi [137]. However, there were several formulations derived from stevia that researchers did not find to be effective. The lack of efficacy was corroborated by Feng and colleagues [68]. It is possible that activity may be preparation-specific, which could explain these divergent findings. However, additional research is needed to further understand the current mixed outcomes. Theophilus and colleagues [169] also showed that when stevia was used with three different antibiotics (doxycycline, cefoperazone, and daptomycin) there was a significant reduction in B. burgdorferi biofilm forms. Safety has been demonstrated at high levels of dietary intake [170,171].

3.1.23. Uncaria tomentosa (Cat’s Claw)

There is evidence that Uncaria tomentosa has antimicrobial effects against human oral pathogens [172]. Research by Feng and colleagues [68] showed in vitro activity against stationary forms of B. burgdorferi, but less impact against the actively growing spirochete form. However, according to Goc and Rath [137], Uncaria tomentosa was shown to be effective against all morphological forms of Borrelia burgdorferi sensu stricto. Published safety data have shown minimal side effects in both animal and human models [173]. Human studies reflected a side effect profile comparable to a placebo in studies ranging from four weeks [174] to 52 weeks [175]. There are reports that Uncaria can have an impact on estrogen binding [176], with potential implications on contraceptive effects [177].

3.1.24. Vitamin C

One peer-reviewed publication described the efficacy of vitamin C against the active, spirochetal forms of Borrelia burgdorferi (and to a lesser extent Borrelia garinii) in vitro [66]. This was supported by another study, which showed additive effects of vitamin C combined with doxycycline against the spirochetal form [178].
Of note, in clinical practice it is recommended that Vitamin C be avoided in combination with hydroxychloroquine due to a possible decrease in hydroxychloroquine efficacy [179].
Furthermore, if supraphysiologic doses of vitamin C are used it is important to rule out G6PD enzyme deficiency due to the risk of potentially triggering hemolysis.
Rizk and colleagues [180] showed that Vitamin C in conjunction with anti-Babesia agent diminazene aceturate (DA) improved the inhibition of Babesia microti growth in vivo. This was initially evaluated in an in vitro model using Babesia bovis and subsequently in an in vivo murine model using Babesia microti. The authors propose this combination as a viable option to decrease drug resistance.

3.1.25. Vitamin D3

The immune system has been established as an important target of vitamin D. The active form of vitamin D (1,25-Dihydroxycholecalciferol [1,25-(OH)2D3]) directly and indirectly suppresses the function of pathogenic T cells while inducing several regulatory T cells that suppress multiple sclerosis and inflammatory bowel disease development [181]. Cantorna and colleagues [182] induced a murine arthritic condition through the injection of collagen or B. burgdorferi. Vitamin D3 supplementation dramatically decreased mouse arthritic symptoms (inflamed and swollen ankles and paws).
According to Goc and colleagues [66,73], Vitamin D has efficacy against the active spirochetal form of Borrelia burgdorferi but not the stationary or biofilm forms [66].

3.2. Combination Protocols Reveal Synergy

In 2017, Goc and colleagues [160] published data on several combinations that showed synergy in vitro against 3 forms of Borrelia (garinii and burgdorferi): active spirochetal, and both stationary and biofilm persister forms. These combinations are reflected in Table 2.
In 2020, Goc and colleagues [183] demonstrated the efficacy and synergy of combination treatment in a Lyme disease animal model and human volunteers. In this study, the researchers used baicalein, luteolin, and rosmarinic acid, along with monolaurin, cis-2-decenoic acid, and iodine/kelp. Inflammatory cytokines such as IL-6, IL-17, TNF-α, and INF-γ were elevated in infected animals, supporting active infection. These inflammatory cytokines normalized in infected animals that were subsequently treated. Furthermore, four weeks of dietary intake of this composition reduced the spirochete burden in animal tissues by about 75%. In a human study of 17 volunteers administered this composition, 67.4% of participants with clinical late or persistent LD (who had not responded to previous antibiotic treatment) responded positively with improved energy status, as well as improved physical and psychological well-being. Additionally, 17.7% had slight improvement, and 17.7% were nonresponsive. In the human observational study, patients received the treatment intervention in the form of capsules containing baicalein 250 mg/day, luteolin 75 mg/day, rosmarinic acid 100 mg/day, monolaurin 250 mg/day, 10-HAD 100 mg/day, and iodine 0.15 mg/day (in the form of kelp) three times per day for six months. These doses represented 1/8th of the minimum inhibitory concentration (MIC) values [183].

4. Miscellaneous

4.1. Synthetic Products

4.1.1. Methylene Blue

Methylene blue (MB) has been used medically for centuries and is best known as an antidote for acquired methemoglobinemia (MetHB) [184]. Methylene blue was the first synthetic antimalarial to be discovered [185], and other common uses include septic shock [186] and as an intraoperative dye [187]. According to Feng and colleagues [104], MB has significant benefits in treating Borrelia burgdorferi stationary persister cells. In addition, Ma and colleagues [82] provided evidence of activity against stationary forms of Bartonella, and Li and colleagues [84] showed that MB had significant effects on the stationary and active log phase forms of Bartonella. Zheng and colleagues [85] reported that MB had relatively low MICs against the log phase form of Bartonella, and was more active than the doxycycline/gentamicin combination against the stationary forms. Lastly, MB and rifampin were the most active agents against the biofilm B. henselae after 6 days of drug exposure. Given its original use as an antimalarial and similarities in pathophysiology and treatment response between Malaria and Babesia [188,189], one could extrapolate that this agent should have anti-Babesia properties. Carvalho and colleagues [190] studied six agents combined with Artusenate in vitro against Babesia bovis and found that the most active drug was MB.

4.1.2. Tetraethylthiuram Disulfide (Disulfiram)

Disulfiram has historically been used as an adjunct to sobriety maintenance in alcoholics. As a repurposed therapy, it has been found to be highly effective as an antiparasitic [191], antifungal [192], and anti-MRSA agent [193], along with having anti-mycobacterium tuberculosis activity [194]. Disulfiram has also been shown to be therapeutic against malaria [195]. Given the clinical and pathophysiological similarities between malaria and the tick-borne pathogen Babesia [188,189], one could propose potential efficacy for Babesia as well.
According to Potula and colleagues [196], disulfiram has excellent borreliacidal activity against both the log and stationary phase B. burgdorferi sensu stricto B31 MI. Their in vivo murine studies showed that disulfiram eliminated the B. burgdorferi sensu stricto B31 MI completely from the hearts and urinary bladder by day 28 post infection. This was also associated with reduced lymphadenopathy and inflammatory markers [196]. In fact, disulfiram was shown to be the most potent of 4366 compounds tested in vitro, with anti B.burgdorferi persister cell activity [197].
In a case series of three patients with Babesia, Liegner [198] was able to show a significant clinical benefit with the use of disulfiram. In a subsequent human study of 71 patients with suspected Lyme disease, 62 of 67 (92.5%) patients treated with disulfiram endorsed a net benefit on their symptoms. Furthermore, 12 of 33 (36.4%) patients who completed one or two courses of “high-dose” therapy (≥4 mg/kg/day vs. a “low-dose” group < 4 mg/kg/day) achieved an “enduring remission”, defined as remaining clinically well for ≥6 months without further anti-infective treatment. Of note, higher doses did produce a higher incidence of adverse reactions including fatigue (66.7%), psychiatric symptoms (48.5%), peripheral neuropathy (27.3%), and the mild to moderate elevation of liver enzymes (15.2%) [199]. To date, there is no evidence in the literature for a therapeutic effect of disulfiram on any Bartonella species.

4.2. Safety

According to Di Lorenzo and colleagues [200], the risk of side effects from botanical medicines is generally felt to be rare. However, each botanical medicine has its own unique action and physiological effect. As with any intervention, there are potentially unforeseen drug interactions or independent idiosyncratic responses [201]. As with any medical therapy, it is recommended that patients work with a knowledgeable practitioner who can advise and guide on the optimal and safest course of therapy. The potential for adverse responses should not preclude the value of the appropriate and judicious use of nutraceuticals, but rather emphasize the need for appropriate oversight. Foundational knowledge of potential interactions and side effects, as well as the use of surveillance labs to identify early adverse hepatic, renal, or bone marrow responses, is recommended.

4.3. Potential Criticisms

This paper attempted to organize the data in a format most useful for decision making at the point of care. However, it must be pointed out that there was heterogeneity in the diagnostic systems identifying pathogens, as well as in the source and potential quality of nutraceutical products being tested. In addition, studies on different pathogen strains were grouped together, and it must be recognized that different strains may respond differently to a given interventions or treatment. For example, with regard to Borrelia, some studies described B. burgdorferi and/or B. garinii strains. Studies on Babesia generally focused on B. microti, but occasionally B. duncani, and in some studies animal strains such as B. gibsoni were evaluated. In addition, most of the conclusions described were derived from in vitro or animal data.
In vitro studies can help assess the potential impact a therapy may have on a pathogen as a direct antimicrobial. However, in vitro studies exist outside the biological organism and thus cannot assess many of the broad pathways through which botanical medicines and nutraceuticals exert their effects. These include anti-inflammatory/anti-cytokine activity, immune system regulation/augmentation, the adaptogenic stimulation of cellular and organismal defense systems, and biofilm disruption, to name a few. Performing in vivo and particularly human research is vital as a future consideration. This would include larger studies evaluating specific interventions and protocols for specific pathogens. Further safety analysis of individual and combined regimens, along with potential drug interactions of both prescriptive and over-the-counter agents, would be of value.

5. Conclusions

In this paper, we reviewed the literature for the broader treatment of tick-borne infections, including Borrelia, Bartonella, and Babesia. By embracing the utility of botanical medicines and nutraceutical agents, our hope is that the field can decrease the need for long-term prescription antibiotics and antimicrobials, while improving overall outcomes. Furthermore, this broader treatment toolkit takes into account the multiple interconnected pathways through which these complex persistent infectious organisms affect the human organism. Ideally, prospective studies will allow for the further expansion of these therapeutic options. As further evidence is generated to support the safety and utility of the interventions discussed, we hope that practitioners can incorporate many of these concepts into their therapeutic strategies. It is our intention to empower clinicians with expanding options, while improving the quality of life and outcomes of our patients.

Author Contributions

Conceptualization, S.M.S.; methodology, S.M.S.; software, S.M.S.; validation, S.M.S. and S.K.S.; writing—original draft preparation, S.M.S.; writing—review and editing, S.M.S. and S.K.S.; supervision, S.M.S.; project administration, S.M.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Not applicable.

Acknowledgments

Lia Gaertner: thank you for your support.

Conflicts of Interest

The lead author is a past president of ILADS (International Lyme and Associated Diseases Society) and the lead author of reference #6, “Chronic Lyme Disease: An Evidence Based Definition by the ILADS Working Group.” The authors otherwise declare no conflict of interests.

Appendix A. Summary of Clinical Impact

Borrelia Bartonella Babesia
AgentIdentified Active Ingredient burgdorferi and/or garinii henselae Species
SpirocheteStationaryBiofilmActiveStationaryBiofilm
Allicin (garlic)Diallyl Disulfide (DADS) 2 16,24
Alchornea cordifoliaEllagic acid 13
Artemisia annua (Sweet wormwood)Artemisinin131,7,11 1,13
Berberis vulgarisBerberine 5 19,25,21
Betula lenta (Birch sweet oil)Methyl salicylate232323
Cinnamomum cassia (Cassia oi)Cinnamaldehyde2,232,3,233,2344 29
Cistus creticus and incanusCarvacrol1,3,17,181,3
Cryptolepis sanguinolentaCryptoleptine1,131 13,3113,31 1,13
Dipsacus sylvestris/Dipsacus fullonum (teasel root )Multiple 6,30
Eugenia caryophyllata (Syzigium aromaticum L. Myrtaceae)- Clove Diallyl Disulfide (DADS) 3344 26,27
Grapefruit see extract (GSE)Flavanoids/ascorbic acid66 4
Juglans nigra (black walnut)Epigallocatechin gallate (EGCG)6613131
KelpIodine101010
Matricaria chamomilla (Chamomile oil German)Chamazulene 232323
Methylene blue Methylene blue 7 5,84,5,887,35
Monolaurin (coconut oil)Lauric acid106,106,10
Nigella sativa (Black cumin)Thymoquinone 36
OreganoCarvacrol & Diallyl Disulfide (DADS)33344
Otoba parvifolia (Banderol)Otoba parvifolia66
Pimenta racemosa (Bay Leaf Oil)Eugonol232323
Piper nigrum (Black Pepper)B-Caryophyllene (BCP) 16
Polygonum cuspidatum (Japanese knotweed)Resveratrol11 3131 1,13
Rhus coriaria L. (Sumac)Multiple 26
Rosmarinic acidDiallyl Disulfide (DADS)10,28
Scutellaria baicalensis (Chineses skullcap)Baicalein1,10,201,10,2010,2 13
SteviaStevioside and rebaudioside6,146,146,14
Tetraethylthiuram Disulfide (Disulfiram)Bis(diethylthiocarbamoyl)disulfide33,3433 34,35
Thymus vulgaris (Thyme oil)Thymol232323
Uncaria tomentosa (Cat’s claw) Isopteropodine & rynchophylline61,66
Vitamin CAscorbic acid10,28
Vitamin D3Cholecalciferol10,28

Appendix B. Reference Key for Appendix A

1. Feng, J.; Leone, J.; Schweig, S.; Zhang, Y. Evaluation of Natural and Botanical Medicines for Activity Against Growing and Non-growing Forms of B. burgdorferi. Front. Med. 2020, 7, 6, https://doi.org/10.3389/fmed.2020.00006.
2. Feng, J.; Shi, W.; Miklossy, J.; Tauxe, G.M.; McMeniman, C.J.; Zhang, Y. Identification of Essential Oils with Strong Activity against Stationary Phase Borrelia burgdorferi. Antibiotics 2018, 7, 89, https://doi.org/10.3390/antibiotics7040089.
3. Feng, J.; Zhang, S.; Shi, W.; Zubcevik, N.; Miklossy, J.; Zhang, Y. Selective Essential Oils from Spice or Culinary Herbs Have High Activity against Stationary Phase and Biofilm Borrelia burgdorferi. Front. Med. 2017, 4, 169–169, https://doi.org/10.3389/fmed.2017.00169.
4. Ma, X.; Shi, W.; Zhang, Y. Essential Oils with High Activity against Stationary Phase Bartonella henselae. Antibiotics 2019, 8, 246, https://doi.org/10.3390/antibiotics8040246.
5. Li, T.; Feng, J.; Xiao, S.; Shi, W.; Sullivan, D.; Zhang, Y. Identification of FDA-Approved Drugs with Activity against Stationary Phase Bartonella henselae. Antibiotics 2019, 8, 50, https://doi.org/10.3390/antibiotics8020050.
6. Goc, A.; Rath, M. The anti-borreliae efficacy of phytochemicals and micronutrients: an update. Ther. Adv. Infect. Dis. 2016, 3, 75–82, https://doi.org/10.1177/2049936116655502.
7. Feng, J.; Weitner, M.; Shi, W.; Zhang, S.; Sullivan, D.; Zhang, Y. Identification of Additional Anti-Persister Activity against Borrelia burgdorferi from an FDA Drug Library. Antibiotics 2015, 4, 397–410, https://doi.org/10.3390/antibiotics4030397.
8. Zheng, X.; Ma, X.; Li, T.; Shi, W.; Zhang, Y. Effect of different drugs and drug combinations on killing stationary phase and biofilms recovered cells of Bartonella henselae in vitro. BMC Microbiol. 2020, 20, 87–9, https://doi.org/10.1186/s12866-020-01777-9.
9. Feng, J.; Wang, T.; Shi, W.; Zhang, S.; Sullivan, D.; Auwaerter, P.G.; Zhang, Y. Identification of novel activity against Borrelia burgdorferi persisters using an FDA approved drug library. Emerg. Microbes Infect. 2014, 3, 1-8, https://doi.org/10.1038/emi.2014.53
10. Goc, A.; Niedzwiecki, A.; Rath, M. In vitro evaluation of antibacterial activity of phytochemicals and micronutrients against Borrelia burgdorferi and Borrelia garinii. J. Appl. Microbiol. 2015, 119, 1561–1572, https://doi.org/10.1111/jam.12970.
11. Feng, J.; Eshi, W.; Ezhang, S.; Esullivan, D.; Auwaerter, P.G.; Ezhang, Y. A Drug Combination Screen Identifies Drugs Active against Amoxicillin-Induced Round Bodies of In Vitro Borrelia burgdorferi Persisters from an FDA Drug Library. Front. Microbiol. 2016, 7, 743, https://doi.org/10.3389/fmicb.2016.00743.
12. Feng, J.; Zhang, S.; Shi, W.; Zhang, Y. Activity of Sulfa Drugs and Their Combinations against Stationary Phase B. burgdorferi In Vitro. 2017, 6, https://doi.org/10.3390/antibiotics6010010.
13. Zhang, Y.; Alvarez-Manzo, H.; Leone, J.; Schweig, S.; Zhang, Y. Botanical Medicines Cryptolepis sanguinolenta, Artemisia annua, Scutellaria baicalensis, Polygonum cuspidatum, and Alchornea cordifolia Demonstrate Inhibitory Activity Against Babesia duncani. Front. Cell. Infect. Microbiol. 2021, 11, https://doi.org/10.3389/fcimb.2021.624745.
14. Theophilus, P.A.S.; Victoria, M.J.; Socarras, K.M.; Filush, K.R.; Gupta, K.; Luecke, D.F.; Sapi, E. Effectiveness of Stevia rebaudiana whole leaf extract against the various morphological forms of Borrelia burgdorferi in vitro. Eur. J. Microbiol. Immunol. 2015, 5, 268–280. https://doi.org/10.1556/1886.2015.00031
15. Luo, J.; Dong, B.; Wang, K.; Cai, S.; Liu, T.; Cheng, X.; Lei, D.; Chen, Y.; Li, Y.; Kong, J.; et al. Baicalin inhibits biofilm formation, attenuates the quorum sensing-controlled virulence and enhances Pseudomonas aeruginosa clearance in a mouse peritoneal implant infection model. PLOS ONE 2017, 12, e0176883, https://doi.org/10.1371/journal.pone.0176883.
16. Zhang, Y.; Bai, C.; Shi, W.; Manzo, H.A.; Zhang, Y. Identification of Essential Oils Including Garlic Oil and Black Pepper Oil with High Activity against Babesia duncani. Pathogens 2020, 9, https://doi.org/10.3390/pathogens9060466.
17. Hutschenreuther, A.; Birkemeyer, C.; Grötzinger, K.; Straubinger, R.; Rauwald, H.W. Growth inhibiting activity of volatile oil from Cistus creticus L. against Borrelia burgdorferi s.s. in vitro. Die Pharm. 2010, 65.
18. Rauwald, H.W.; Liebold, T.; Grötzinger, K.; Lehmann, J.; Kuchta, K. Labdanum and Labdanes of Cistus creticus and C. ladanifer: Anti-Borrelia activity and its phytochemical profiling. Phytomedicine 2019, 60, 152977, https://doi.org/10.1016/j.phymed.2019.152977.
19. Batiha, G.E.-S.; Magdy Beshbishy, A.; Adeyemi, O.S.; Nadwa, E.H.; Rashwan, E.K.M.; Alkazmi, L.M.; Elkelish, A.A.; Igarashi, I. Phytochemical Screening and Antiprotozoal Effects of the Methanolic Berberis Vulgaris and Acetonic Rhus Coriaria Extracts. Molecules 2020, 25, 550, https://doi.org/10.3390/molecules25030550.
20. Goc, A.; Niedzwiecki, A.; Rath, M. Reciprocal cooperation of phytochemicals and micronutrients against typical and atypical forms of Borrelia sp. J. Appl. Microbiol. 2017, 123, 637–650, https://doi.org/10.1111/jam.13523.
21. Elkhateeb, A.; Yamada, K.; Takahashi, K.; Matsuura, H.; Yamasaki, M.; Maede, Y.; Katakura, K.; Nabeta, K. Anti-Babesial Compounds from Berberis Vulgaris. Nat. Prod. Commun. 2007, 2, https://doi.org/10.1177/1934578x0700200213.
22. Brorson, O.; Brorson, S.-H. Grapefruit Seed Extract is a Powerful in vitro Agent Against Motile and Cystic Forms of Borrelia burgdorferi sensu lato. Infection 2007, 35, 206–208, https://doi.org/10.1007/s15010-007-6105-0.
23. Goc, A.; Niedzwiecki, A.; Rath, M. Anti-borreliae efficacy of selected organic oils and fatty acids. BMC Complement. Altern. Med. 2019, 19, 1–11, https://doi.org/10.1186/s12906-019-2450-7.
24. Salama, A.A.; AbouLaila, M.; Terkawi, M.A.; Mousa, A.; El-Sify, A.; Allaam, M.; Zaghawa, A.; Yokoyama, N.; Igarashi, I. Inhibitory effect of allicin on the growth of Babesia and Theileria equi parasites. Parasitol. Res. 2013, 113, 275–283, https://doi.org/10.1007/s00436-013-3654-2.
25. Subeki; Matsuura, H.; Takahashi, K.; Yamasaki, M.; Yamato, O.; Maede, Y.; Katakura, K.; Suzuki, M.; Trimurningsih; Chairul; et al. Antibabesial Activity of Protoberberine Alkaloids and 20-Hydroxyecdysone from Arcangelisia flava against Babesia gibsoni in Culture. J. Veter- Med Sci. 2005, 67, 223–227, https://doi.org/10.1292/jvms.67.223.
26. Batiha, G.E.-S.; Beshbishy, A.M.; El-Mleeh, A.; Abdel-Daim, M.M.; Devkota, H.P. Traditional Uses, Bioactive Chemical Constituents, and Pharmacological and Toxicological Activities of Glycyrrhiza glabra L. (Fabaceae). Biomolecules 2020, 10, 352, https://doi.org/10.3390/biom10030352.
27. Batiha, G.E.-S.; Beshbishy, A.M.; Tayebwa, D.S.; Shaheen, H.M.; Yokoyama, N.; Igarashi, I. Inhibitory effects of Syzygium aromaticum and Camellia sinensis methanolic extracts on the growth of Babesia and Theileria parasites. Ticks Tick-borne Dis. 2019, 10, 949–958, https://doi.org/10.1016/j.ttbdis.2019.04.016.
28. Goc, A.; Niedzwiecki, A.; Rath, M. Cooperation of Doxycycline with Phytochemicals and Micronutrients Against Active and Persistent Forms of Borrelia sp. Int. J. Biol. Sci. 2016, 12, 1093–1103, https://doi.org/10.7150/ijbs.16060.
29. Batiha, G.E.-S.; Beshbishy, A.M.; Guswanto, A.; Nugraha, A.; Munkhjargal, T.; Abdel-Daim, M.M.; Mosqueda, J.; Igarashi, I. Phytochemical Characterization and Chemotherapeutic Potential of Cinnamomum verum Extracts on the Multiplication of Protozoan Parasites In Vitro and In Vivo. Molecules 2020, 25, 996, https://doi.org/10.3390/molecules25040996.
30. Rauwald, H.W.; Liebold, T.; Straubinger, R.K. Growth inhibiting activity of lipophilic extracts from Dipsacus sylvestris Huds. roots against Borrelia burgdorferi s. s. in vitro. 2011, 628–630, https://doi.org/10.1691/PH.2011.0887.
31. Ma, X.; Leone, J.; Schweig, S.; Zhang, Y. Botanical Medicines With Activity Against Stationary Phase Bartonella henselae. Infect. Microbes Dis. 2021, 3, 158–167, https://doi.org/10.1097/im9.0000000000000069.
32. Rizk, M.A.; El-Sayed, S.A.E.-S.; Igarashi, I. Ascorbic acid co-administration with a low dose of diminazene aceturate inhibits the in vitro growth of Theileria equi, and the in vivo growth of Babesia microti. Parasitol. Int. 2022, 90, https://doi.org/10.1016/j.parint.2022.102596.
33. Potula, H.-H.S.K.; Shahryari, J.; Inayathullah, M.; Malkovskiy, A.V.; Kim, K.-M.; Rajadas, J. Repurposing Disulfiram (Tetraethylthiuram Disulfide) as a Potential Drug Candidate against Borrelia burgdorferi In Vitro and In Vivo. Antibiotics 2020, 9, 633, https://doi.org/10.3390/antibiotics9090633.
34. Liegner, K.B. Disulfiram (Tetraethylthiuram Disulfide) in the Treatment of Lyme Disease and Babesiosis: Report of Experience in Three Cases. Antibiotics 2019, 8, 72, https://doi.org/10.3390/antibiotics8020072.
35. Carvalho, L.J.M.; Tuvshintulga, B.; Nugraha, A.B.; Sivakumar, T.; Yokoyama, N. Activities of artesunate-based combinations and tafenoquine against Babesia bovis in vitro and Babesia microti in vivo. Parasites Vectors 2020, 13, 1–9, https://doi.org/10.1186/s13071-020-04235-7.
36. El-Sayed, S.A.E.-S.; Rizk, M.A.; Yokoyama, N.; Igarashi, I. Evaluation of the in vitro and in vivo inhibitory effect of thymoquinone on piroplasm parasites. Parasites Vectors 2019, 12, 37, https://doi.org/10.1186/s13071-019-3296-z.

References

  1. Schwartz, A.M.; Hinckley, A.F.; Mead, P.S.; Hook, S.A.; Kugeler, K.J. Surveillance for Lyme Disease—United States, 2008–2015. MMWR Surveill. Summ. 2017, 66, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Gray, J.S.; Kirstein, F.; Robertson, J.N.; Stein, J.; Kahl, O. Borrelia burgdorferi sensu lato in Ixodes ricinus Ticks and Rodents in a Recreational Park in South-Western Ireland. Exp. Appl. Acarol. 1999, 23, 717–729. [Google Scholar] [CrossRef] [PubMed]
  3. Schwartz, A.M.; Kugeler, K.J.; Nelson, C.A.; Marx, G.E.; Hinckley, A.F. Evaluation of Commercial Insurance Claims as an Annual Data Source for Lyme Disease Diagnoses. Emerg. Infect. Dis. 2021, 27, 499–507. [Google Scholar] [CrossRef]
  4. Kugeler, K.J.; Schwartz, A.M.; Delorey, M.J.; Mead, P.S.; Hinckley, A.F. Estimating the Frequency of Lyme Disease Diagnoses—United States, 2010–2018. Emerg. Infect. Dis. 2021, 27, 616–619. [Google Scholar] [CrossRef]
  5. Aucott, J.N.; Rebman, A.W.; Crowder, L.A.; Kortte, K.B. Post-Treatment Lyme Disease Syndrome Symptomatology and the Impact on Life Functioning: Is There Something Here? Qual. Life Res. 2013, 22, 75–78. [Google Scholar] [CrossRef] [Green Version]
  6. Shor, S.; Green, C.; Szantyr, B.; Phillips, S.; Liegner, K.; Burrascano, J.J., Jr.; Bransfield, R.; Maloney, E.L. Chronic Lyme Disease: An Evidence-Based Definition by the ILADS Working Group. Antibiotics 2019, 8, 269. [Google Scholar] [CrossRef] [Green Version]
  7. Berger, B.W. Dermatologic Manifestations of Lyme Disease. Rev. Infect. Dis. 1989, 11 (Suppl. 6), S1475–S1481. [Google Scholar] [CrossRef] [PubMed]
  8. Preac-Mursic, V.; Pfister, H.W.; Spiegel, H.; Burk, R.; Wilske, B.; Reinhardt, S.; Böhmer, R. First Isolation of Borrelia burgdorferi from an Iris Biopsy. J. Clin. Neuroophthalmol. 1993, 13, 155–161. [Google Scholar]
  9. Schmidli, J.; Hunziker, T.; Moesli, P.; Schaad, U.B. Cultivation of Borrelia burgdorferi from Joint Fluid Three Months after Treatment of Facial Palsy Due to Lyme Borreliosis. J. Infect. Dis. 1988, 158, 905–906. [Google Scholar] [CrossRef] [Green Version]
  10. Kirsch, M.; Ruben, F.L.; Steere, A.C.; Duray, P.H.; Norden, C.W.; Winkelstein, A. Fatal Adult respiratory distress syndrome in a patient with Lyme disease. JAMA 1988, 259, 2737–2739. [Google Scholar] [CrossRef]
  11. Preac-Mursic, V.; Weber, K.; Pfister, H.W.; Wilske, B.; Gross, B.; Baumann, A.; Prokop, J. Survival of Borrelia burgdorferi in antibiotically treated patients with Lyme borreliosis. Infection 1989, 17, 355–359. [Google Scholar] [CrossRef] [PubMed]
  12. Pfister, H.W.; Preac-Mursic, V.; Wilske, B.; Schielke, E.; Sorgel, F.; Einhaupl, K.M. Randomized comparison of ceftriaxone and cefotaxime in Lyme neuroborreliosis. J. Infect. Dis. 1991, 163, 311–318. [Google Scholar] [CrossRef] [PubMed]
  13. Liegner, K.B.; Shapiro, J.R.; Ramsay, D.; Halperin, A.; Hogrefe, W.; Kong, L. Recurrent erythema migrans despite extended antibiotic treatment with minocycline in a patient with persisting Borrelia burgdorferi infection. J. Am. Acad. Dermatol. 1993, 28 (Pt 2), 312–314. [Google Scholar] [CrossRef]
  14. Strle, F.; Preac-Mursic, V.; Cimperman, J.; Ruzic, E.; Maraspin, V.; Jereb, M. Azithromycin versus doxycycline for treatment of erythema migrans: Clinical and microbiological findings. Infection 1993, 21, 83–88. [Google Scholar] [CrossRef] [PubMed]
  15. Weber, K.; Wilske, B.; Preac-Mursic, V.; Thurmayr, R. Azithromycin versus penicillin V for the treatment of early Lyme borreliosis. Infection 1993, 21, 367–372. [Google Scholar] [CrossRef] [PubMed]
  16. Battafarano, D.F.; Combs, J.A.; Enzenauer, R.J.; Fitzpatrick, J.E. Chronic septic arthritis caused by Borrelia burgdorferi. Clin. Orthop. Relat. Res. 1993, 297, 238–241. [Google Scholar] [CrossRef]
  17. Chancellor, M.B.; McGinnis, D.E.; Shenot, P.J.; Kiilholma, P.; Hirsch, I.H. Urinary dysfunction in Lyme disease. J. Urol. 1993, 149, 26–30. [Google Scholar] [CrossRef]
  18. Bradley, J.F.; Johnson, R.C.; Goodman, J.L. The persistence of spirochetal nucleic acids in active Lyme arthritis. Ann. Intern. Med. 1994, 120, 487–489. [Google Scholar] [CrossRef]
  19. Lawrence, C.; Lipton, R.B.; Lowy, F.D.; Coyle, P.K. Seronegative chronic relapsing neuroborreliosis. Eur. Neurol. 1995, 35, 113–117. [Google Scholar] [CrossRef]
  20. Strle, F.; Maraspin, V.; Lotric-Furlan, S.; Ruzic-Sabljic, E.; Cimperman, J. Azithromycin and doxycycline for treatment of Borrelia culture-positive erythema migrans. Infection 1996, 24, 64–68. [Google Scholar] [CrossRef]
  21. Oksi, J.; Nikoskelainen, J.; Viljanen, M.K. Comparison of oral cefixime and intravenous ceftriaxone followed by oral amoxicillin in disseminated Lyme borreliosis. Eur. J. Clin. Microbiol. Infect. Dis. 1998, 17, 715–719. [Google Scholar] [CrossRef]
  22. Priem, S.; Burmester, G.R.; Kamradt, T.; Wolbart, K.; Rittig, M.G.; Krause, A. Detection of Borrelia burgdorferi by polymerase chain reaction in synovial membrane, but not in synovial fluid from patients with persisting Lyme arthritis after antibiotic therapy. Ann. Rheum. Dis. 1998, 57, 118–121. [Google Scholar] [CrossRef]
  23. Hudson, B.J.; Stewart, M.; Lennox, V.A.; Fukunaga, M.; Yabuki, M.; Macorison, H.; Kitchener-Smith, J. Culture-positive Lyme borreliosis. Med. J. Aust. 1998, 168, 500–502. [Google Scholar] [CrossRef] [PubMed]
  24. Oksi, J.; Marjamaki, M.; Nikoskelainen, J.; Viljanen, M.K. Borrelia burgdorferi detected by culture and PCR in clinical relapse of disseminated Lyme borreliosis. Ann. Med. 1999, 31, 225–232. [Google Scholar] [CrossRef]
  25. Breier, F.; Khanakah, G.; Stanek, G.; Kunz, G.; Aberer, E.; Schmidt, B.; Tappeiner, G. Isolation and polymerase chain reaction typing of Borrelia afzelii from a skin lesion in a seronegative patient with generalized ulcerating bullous lichen sclerosus et atrophicus. Br. J. Dermatol. 2001, 144, 387–392. [Google Scholar] [CrossRef] [PubMed]
  26. Hunfeld, K.P.; Ruzic-Sabljic, E.; Norris, D.E.; Kraiczy, P.; Strle, F. In vitro susceptibility testing of Borrelia burgdorferi sensu lato isolates cultured from patients with erythema migrans before and after antimicrobial chemotherapy. Antimicrob. Agents Chemother. 2005, 49, 1294–1301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Marques, A.; Telford, S.R.; Turk, S.P.; Chung, E.; Williams, C.; Dardick, K.; Krause, P.J.; Brandeburg, C.; Crowder, C.D.; Carolan, H.E.; et al. Xenodiagnosis to detect Borrelia burgdorferi infection: A first-in-human study. Clin. Infect. Dis. 2014, 58, 937–945. [Google Scholar] [CrossRef]
  28. Embers, M.E.; Barthold, S.W.; Borda, J.T.; Bowers, L.; Doyle, L.; Hodzic, E.; Jacobs, M.B.; Hasenkampf, N.R.; Martin, D.S.; Narasimhan, S.; et al. Persistence of Borrelia burgdorferi in Rhesus Macaques following antibiotic treatment of disseminated infection. PLoS ONE 2012, 7, e29914. [Google Scholar] [CrossRef]
  29. Hodzic, E.; Feng, S.; Holden, K.; Freet, K.J.; Barthold, S.W. Persistence of Borrelia burgdorferi following antibiotic treatment in mice. Antimicrob. Agents Chemother. 2008, 52, 1728–1736. [Google Scholar] [CrossRef] [Green Version]
  30. Barthold, S.W.; Hodzic, E.; Imai, D.M.; Feng, S.; Yang, X.; Luft, B.L. Ineffectiveness of tigecycline against persistent Borrelia burgdorferi. Antimicrob. Agents Chemother. 2010, 54, 643–651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Straubinger, R.K.; Summers, B.A.; Chang, Y.F.; Appel, M.J. Persistence of Borrelia burgdorferi in experimentally infected dogs after antibiotic treatment. J. Clin. Microbiol. 1997, 35, 111–116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Embers, M.E.; Hasenkampf, N.R.; Jacobs, M.B.; Tardo, A.C.; Doyle-Meyers, L.A.; Philipp, M.T.; Hodzic, E. Variable manifestations, diverse seroreactivity and post-treatment persistence in non-human primates exposed to Borrelia burgdorferi by tick feeding. PLoS ONE 2017, 12, e0189071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Rudenko, N.; Golovchenko, M.; Kybicova, K.; Vancova, M. Metamorphoses of Lyme disease spirochetes: Phenomenon of Borrelia persisters. Parasites Vectors 2019, 12, 237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Brorson, O.; Brorson, S.H. In vitro conversion of Borrelia burgdorferi to cystic forms in spinal fluid, and transformation to mobile spirochetes by incubation in BSK-H medium. Infection 1998, 26, 144–150. [Google Scholar] [CrossRef]
  35. Murgia, R.; Cinco, M. Induction of cystic forms by different stress conditions in Borrelia burgdorferi. APMIS 2004, 112, 57–62. [Google Scholar] [CrossRef]
  36. Sapi, E.; Bastian, S.L.; Mpoy, C.M.; Scott, S.; Rattelle, A.; Pabbati, N.; Poruri, A.; Burugu, D.; Theophilus, P.A.; Pham, T.V.; et al. Characterization of biofilm formation by Borrelia burgdorferi in vitro. PLoS ONE 2012, 7, e48277. [Google Scholar] [CrossRef]
  37. Sapi, E.; Kaur, N.; Anyanwu, S.; Luecke, D.F.; Datar, A.; Patel, S.; Rossi, M.; Stricker, R.B. Evaluation of in-vitro antibiotic susceptibility of different morphological forms of Borrelia burgdorferi. Infect. Drug Resist. 2011, 4, 97–113. [Google Scholar]
  38. Sharma, B.; Brown, A.V.; Matluck, N.E.; Hu, L.T.; Lewis, K. Borrelia burgdorferi, the causative agent of Lyme disease, forms drug-tolerant persister cells. Antimicrob. Agents Chemother. 2015, 59, 4616–4624. [Google Scholar] [CrossRef] [Green Version]
  39. Feng, J.; Auwaerter, P.G.; Zhang, Y. Drug Combinations against Borrelia burgdorferi persisters in vitro: Eradication achieved by using daptomycin, cefoperazone and doxycycline. PLoS ONE 2015, 10, e0117207. [Google Scholar] [CrossRef] [Green Version]
  40. Caskey, J.R.; Embers, M.E. Persister development by Borrelia burgdorferi populations in vitro. Antimicrob. Agents Chemother. 2015, 59, 6288–6295. [Google Scholar] [CrossRef] [Green Version]
  41. Feng, J.; Wang, T.; Shi, W.; Zhang, S.; Sullivan, D.; Auwaerter, P.G.; Zhang, Y. Identification of novel activity against Borrelia burgdorferi persisters using an FDA approved drug library. Emerg. Microbes Infect. 2014, 3, e49. [Google Scholar] [CrossRef] [PubMed]
  42. Sapi, E.; Balasubramanian, K.; Poruri, A.; Maghsoudlou, J.S.; Socarras, K.M.; Timmaraju, A.V.; Filush, K.R.; Gupta, K.; Shaikh, S.; Theophilus, P.A.; et al. Evidence of in vivo existence of Borrelia biofilm in Borrelial lymphocytomas. Eur. J. Microbiol. Immunol. 2016, 6, 9–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Sanchez-Vicente, S.; Tagliafierro, T.; Coleman, J.L.; Benach, J.L.; Tokarz, R. Polymicrobial Nature of Tick-Borne Diseases. mBio 2019, 10, e02055-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Garcia-Monco, J.C.; Benach, J.L. Lyme Neuroborreliosis: Clinical Outcomes, Controversy, Pathogenesis, and Polymicrobial Infections. Ann. Neurol. 2019, 85, 21–31. [Google Scholar] [CrossRef]
  45. Citera, M.; Freeman, P.R.; Horowitz, R.I. Empirical validation of the Horowitz Multiple Systemic Infectious Disease Syndrome Questionnaire for suspected Lyme disease. Int. J. Gen. Med. 2017, 10, 249–273. [Google Scholar] [CrossRef] [Green Version]
  46. Horowitz, R.I.; Freeman, P.R. Precision Medicine: The Role of the MSIDS Model in Defining, Diagnosing, and Treating Chronic Lyme Disease/Post Treatment Lyme Disease Syndrome and Other Chronic Illness: Part 2. Healthcare 2018, 6, 129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Krause, P.J. Human babesiosis. Int. J. Parasitol. 2019, 49, 165–174. [Google Scholar] [CrossRef]
  48. Dunn, J.M.; Krause, P.J.; Davis, S.; Vannier, E.G.; Fitzpatrick, M.C.; Rollend, L.; Belperron, A.A.; States, S.L.; Stacey, A.; Bockenstedt, L.K.; et al. Borrelia burgdorferi promotes the establishment of Babesia microti in the northeastern United States. PLoS ONE 2014, 9, e115494. [Google Scholar]
  49. Madison-Antenucci, S.; Kramer, L.D.; Gebhardt, L.L.; Kauffman, E. Emerging Tick-Borne Diseases. Clin. Microbiol. Rev. 2020, 33, e00083-18. [Google Scholar] [CrossRef]
  50. de la Fuente, J.; Antunes, S.; Bonnet, S.; Cabezas-Cruz, A.; Domingos, A.G.; Estrada-Peña, A.; Johnson, N.; Kocan, K.M.; Mansfield, K.L.; Nijhof, A.M.; et al. Tick-Pathogen Interactions and Vector Competence: Identification of Molecular Drivers for Tick-Borne Diseases. Front. Cell. Infect. Microbiol. 2017, 7, 114. [Google Scholar] [CrossRef] [Green Version]
  51. Krause, P.J.; Telford, S.R.; Spielman, A.; Sikand, V.; Ryan, R.; Christianson, D.; Burke, G.; Brassard, P.; Pollack, R.; Peck, J.; et al. Concurrent Lyme disease and babesiosis. Evidence for increased severity and duration of illness. JAMA 1996, 275, 1657–1660. [Google Scholar] [CrossRef] [PubMed]
  52. Wormser, G.P.; Dattwyler, R.J.; Shapiro, E.D.; Halperin, J.J.; Steere, A.C.; Klempner, M.S.; Krause, P.J.; Bakken, J.S.; Strle, F.; Stanek, G.; et al. The Clinical Assessment, Treatment and Prevention of Lyme Disease, Human Granulocytic Anaplasmosis, and Babebiosis: Clinical Practice Guidelines by the Infectious Disease Society of America. Clin. Infect. Dis. 2006, 43, 1089–1134. [Google Scholar] [CrossRef] [PubMed]
  53. Lantos, P.M.; Rumbaugh, J.; Bockenstedt, L.K.; Falck-Ytter, Y.T.; Aguero-Rosenfeld, M.E.; Auwaerter, P.G.; Baldwin, K.; Bannuru, R.R.; Belani, K.K.; Bowie, W.R.; et al. Clinical Practice Guidelines by the Infectious Diseases Society of America (IDSA), American Academy of Neurology (AAN), and American College of Rheumatology (ACR): 2020 Guidelines for the Prevention, Diagnosis, and Treatment of Lyme Disease. Arthritis Rheumatol. 2021, 73, 12–20. [Google Scholar] [CrossRef] [PubMed]
  54. Cameron, D.J.; Johnson, L.B.; Maloney, E.L. Evidence assessments and guideline recommendations in Lyme disease: The clinical management of known tick bites, erythema migrans rashes and persistent disease. Expert. Rev. Anti Infect. Ther. 2014, 12, 1103–1135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Klempner, M.S.; Hu, L.T.; Evans, J.; Schmid, C.H.; Johnson, G.M.; Trevino, R.P.; Norton, D.; Levy, L.; Wall, D.; McCall, J.; et al. Two controlled trials of antibiotic treatment in patients with persistent symptoms and a history of Lyme disease. N. Engl. J. Med. 2001, 345, 85–92. [Google Scholar] [CrossRef] [Green Version]
  56. Fallon, B.A.; Keilp, J.G.; Corbera, K.M.; Petkova, E.; Britton, C.B.; Dwyer, E.; Slavov, I.; Cheng, J.; Dobkin, J.; Nelson, D.R.; et al. A randomized, placebo-controlled trial of repeated IV antibiotic therapy for Lyme encephalopathy. Neurology 2008, 70, 992–1003. [Google Scholar] [CrossRef] [Green Version]
  57. Krupp, L.B.; Hyman, L.G.; Grimson, R.; Coyle, P.K.; Melville, P.; Ahnn, S.; Dattwyler, R.; Chandler, B. Study and treatment of post Lyme disease (STOP-LD): A randomized double masked clinical trial. Neurology 2003, 60, 1923–1930. [Google Scholar] [CrossRef]
  58. DeLong, A.K.; Blossom, B.; Maloney, E.L.; Phillips, S.E. Antibiotic retreatment of Lyme disease: Review of randomized, placebo-controlled, clinical trials. Contemp. Clin. Trials 2012, 33, 1132–1142. [Google Scholar] [CrossRef]
  59. Cameron, D.J. Generalizability in two clinical trials of Lyme disease. Epidemiol. Perspect. Innov. 2006, 3, 12. [Google Scholar] [CrossRef] [Green Version]
  60. Stricker, R.B. Counterpoint: Long-term antibiotic therapy improves persistent symptoms associated with Lyme disease. Clin. Infect. Dis. 2007, 45, 149–157. [Google Scholar] [CrossRef]
  61. Nahid, P.; Mase, S.R.; Migliori, G.B.; Sotgiu, G.; Bothamley, G.H.; Brozek, J.L.; Cattamanchi, A.; Cegielski, J.P.; Chen, L.; Daley, C.L.; et al. Treatment of Drug-Resistant Tuberculosis. An Official ATS/CDC/ERS/IDSA Clinical Practice Guideline. Am. J. Respir. Crit. Care Med. 2019, 200, e93–e142. [Google Scholar] [CrossRef] [PubMed]
  62. Bartos, G.; Sheuring, R.; Combs, A.; Rivlin, D. Treatment of histoid leprosy: A lack of consensus. Int. J. Dermatol. 2020, 59, 1264–1269. [Google Scholar] [CrossRef] [PubMed]
  63. Million, M.; Thuny, F.; Richet, H.; Raoult, D. Long-term outcome of Q fever endocarditis: A 26-year personal survey. Lancet Infect. Dis. 2010, 10, 527–535. [Google Scholar] [CrossRef] [PubMed]
  64. Horowitz, R.I.; Murali, K.; Gaur, G.; Freeman, P.R.; Sapi, E. Effect of dapsone alone and in combination with intracellular antibiotics against the biofilm form of B. burgdorferi. BMC Res. Notes. 2020, 13, 455. [Google Scholar] [CrossRef]
  65. Cowan, M.M. Plant products as antimicrobial agents. Clin. Microbiol. Rev. 1999, 12, 564–582. [Google Scholar] [CrossRef] [Green Version]
  66. Goc, A.; Niedzwiecki, A.; Rath, M. In vitro evaluation of antibacterial activity of phytochemicals and micronutrients against Borrelia burgdorferi and Borrelia garinii. J. Appl. Microbiol. 2015, 119, 1561–1572. [Google Scholar] [CrossRef] [Green Version]
  67. Goc, A.; Niedzwiecki, A.; Rath, M. Anti-borreliae efficacy of selected organic oils and fatty acids. BMC Complement. Altern. Med. 2019, 19, 40. [Google Scholar] [CrossRef]
  68. Feng, J.; Leone, J.; Schweig, S.; Zhang, Y. Evaluation of Natural and Botanical Medicines for Activity Against Growing and Non-growing Forms of B. burgdorferi. Front. Med. 2020, 7, 6. [Google Scholar] [CrossRef] [Green Version]
  69. Nazzaro, F.; Fratianni, F.; De Martino, L.; Coppola, R.; De Feo, V. Effect of essential oils on pathogenic bacteria. Pharmaceuticals 2013, 6, 1451–1474. [Google Scholar] [CrossRef]
  70. Chorianopoulos, N.G.; Giaouris, E.D.; Skandamis, P.N.; Haroutounian, S.A.; Nychas, G.J.E. Disinfectant test against monoculture and mixed-culture biofilms composed of technological, spoilage and pathogenic bacteria: Bactericidal effect of essential oil and hydrosol of Satureja thymbra and comparison with standard acid-base sanitizers. J. Appl. Microbiol. 2008, 104, 1586–1599. [Google Scholar] [CrossRef]
  71. Burt, S.A.; Reinders, R.D. Antibacterial activity of selected plant essential oils against Escherichia coli O157:H7. Lett. Appl. Microbiol. 2003, 36, 162–167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. De Martino, L.; de Feo, V.; Nazzaro, F. Chemical composition and in vitro antimicrobial and mutagenic activities of seven Lamiaceae essential oils. Molecules 2009, 14, 4213–4230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Feng, J.; Zhang, S.; Shi, W.; Zubcevik, N.; Miklossy, J.; Zhang, Y. Selective Essential Oils from Spice or Culinary Herbs Have High Activity against Stationary Phase and Biofilm Borrelia burgdorferi. Front. Med. 2017, 4, 169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Zhang, Y.; Alvarez-Manzo, H.; Leone, J.; Schweig, S.; Zhang, Y. Botanical Medicines Cryptolepis sanguinolenta, Artemisia annua, Scutellaria baicalensis, Polygonum cuspidatum, and Alchornea cordifolia Demonstrate Inhibitory Activity Against Babesia duncani. Front. Cell. Infect. Microbiol. 2021, 11, 624745. [Google Scholar] [CrossRef] [PubMed]
  75. Zhai, X.; Li, L.; Zhang, P.; Guo, Y.; Jiang, H.; He, W.; Li, Y.; Zhang, B.; Yao, D. Evaluation of The Inhibitory Effects of Six Natural Product Extracts Against Babesia Gibsoni In Vitro and In Vivo. J. Parasitol. 2022, 108, 301–305. [Google Scholar] [CrossRef] [PubMed]
  76. Batiha, G.E.; Beshbishy, A.M.; Guswanto, A.; Nugraha, A.; Munkhjargal, T.; MAbdel-Daim, M.; Mosqueda, J.; Igarashi, I. Phytochemical Characterization and Chemotherapeutic Potential of Cinnamomum verum Extracts on the Multiplication of Protozoan Parasites In Vitro and In Vivo. Molecules 2020, 25, 996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Zhang, Y.; Bai, C.; Shi, W.; Alvarez-Manzo, H.; Zhang, Y. Identification of Essential Oils Including Garlic Oil and Black Pepper Oil with High Activity Against Babesia duncani. Pathogens 2020, 9, 466. [Google Scholar] [CrossRef]
  78. Chmielewski, T.; Kuśmierczyk, M.; Fiecek, B.; Roguska, U.; Lewandowska, G.; Parulski, A.; Cielecka-Kuszyk, J.; Tylewska-Wierzbanowska, S. Tick-borne pathogens Bartonella spp., Borrelia burgdorferi sensu lato, Coxiella burnetii and Rickettsia spp. may trigger endocarditis. Adv. Clin. Exp. Med. 2019, 28, 937–943. [Google Scholar] [CrossRef] [Green Version]
  79. Maggi, R.G.; Mozayeni, B.R.; Pultorak, E.L.; Hegarty, B.C.; Bradley, J.M.; Correa, M.; Breitschwerdt, E.B. Bartonella spp. Bacteremia and Rheumatic Symptoms in Patients from Lyme Disease-Endemic Region. Emerg. Infect. Dis. 2012, 18, 783–791. [Google Scholar] [CrossRef]
  80. Jensen, B.B.; Ocias, L.F.; Andersen, N.S.; Dessau, R.B.; Krogfelt, K.A.; Skarphedinsson, S. Tick-borne infections in Denmark. Ugeskr. Laeger. 2017, 179, V01170027. [Google Scholar]
  81. Angelakis, E.; Raoult, D. Pathogenicity and treatment of Bartonella infections. Int. J. Antimicrob. Agents 2014, 44, 16–25. [Google Scholar] [CrossRef] [PubMed]
  82. Ma, X.; Shi, W.; Zhang, Y. Essential Oils with High Activity against Stationary Phase Bartonella henselae. Antibiotics 2019, 8, 246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Scutera, S.; Mitola, S.; Sparti, R.; Salvi, V.; Grillo, E.; Piersigilli, G.; Bugatti, M.; Alotto, D.; Schioppa, T.; Sozzani, S.; et al. Bartonella henselae Persistence within Mesenchymal Stromal Cells Enhances Endothelial Cell Activation and Infectibility That Amplifies the Angiogenic Process. Infect. Immun. 2021, 89, e0014121. [Google Scholar] [CrossRef] [PubMed]
  84. Li, T.; Feng, J.; Xiao, S.; Shi, W.; Sullivan, D.; Zhang, Y. Identification of FDA-Approved Drugs with Activity against Stationary Phase Bartonella henselae. Antibiotics 2019, 8, 50. [Google Scholar] [CrossRef] [Green Version]
  85. Zheng, X.; Ma, X.; Li, T.; Shi, W.; Zhang, Y. Effect of Different Drugs and Drug Combinations on Killing Stationary Phase and Biofilms Recovered Cells of Bartonella Henselae In Vitro. BMC Microbiol. 2020, 20, 87. [Google Scholar] [CrossRef]
  86. Ebi, G.C. Antimicrobial activities of Alchornea cordifolia. Fitoterapia 2001, 72, 69–72. [Google Scholar] [CrossRef]
  87. Manga, H.M.; Brkic, D.; Marie, D.E.; Quetin-Leclercq, J. In vivo anti-inflammatory activity of Alchornea cordifolia (Schumach. Thonn.) Mull. Arg. (Euphorbiaceae). J. Ethnopharmacol. 2004, 92, 209–214. [Google Scholar] [CrossRef]
  88. Mustofa, A.; Benoit-Vical, F.; Pelissier, Y.; Kone-Bamba, D.; Mallie, M. Antiplasmodial Activity of Plant Extracts Used in West African Traditional Medicine. J. Ethnopharmacol. 2000, 73, 145–151. [Google Scholar]
  89. Mesia, G.K.; Tona, G.L.; Nanga, T.H.; Cimanga, R.K.; Apers, S.; Cos, P.; Maes, L.; Pieters, L.; Vlietinck, A.J. Antiprotozoal Cytotoxic Screening of 45 Plant extracts from Democratic Republic of Congo. J. Ethnopharmacol. 2008, 115, 409–415. [Google Scholar] [CrossRef]
  90. Ayisi, N.K.; Appiah-Opong, R.; Gyan, B.; Bugyei, K.; Ekuban, F. Plasmodium falciparum: Assessment of Selectivity of Action of Chloroquine, Alchornea cordifolia, Ficus polita, and Other Drugs by a Tetrazolium-Based Colorimetric Assay. Malar. Res. Treat 2011, 2011, 816250. [Google Scholar] [CrossRef] [Green Version]
  91. Boniface, P.K.; Ferreira, S.B.; Kaiser, C.R. Recent Trends in Phytochemistry, Ethnobotany and Pharmacological Significance of Alchornea cordifolia (Schumach. & Thonn.) Muell. Arg. J. Ethnopharmacol. 2016, 191, 216–244. [Google Scholar]
  92. Gatsing, D.; Nkeugouapi, C.F.N.; Nji-Nkah, B.F.; Kuiate, J.R.; Tchouanguep, F.M. Antibacterial Activity, Bioavailability and Acute Toxicity Evaluation of the Leaf Extract of Alchornea cordifolia (Euphorbiaceae). Int. J. Pharmacol. 2010, 6, 173–182. [Google Scholar] [CrossRef] [Green Version]
  93. Ajibade, T.O.; Olayemi, F.O. Reproductive and toxic effects of methanol extract of Alchornea cordifolia leaf in male rats. Andrologia 2015, 47, 1034–1040. [Google Scholar] [CrossRef]
  94. Djimeli, M.N.; Fodouop, S.P.C.; Njateng, G.S.S.; Fokunang, C.; Tala, D.S.; Kengni, F.; Gatsing, D. Antibacterial activities and toxicological study of the aqueous extract from leaves of Alchornea cordifolia (Euphorbiaceae). BMC Complement. Altern. Med. 2017, 17, 349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Ankri, S.; Mirelman, D. Antimicrobial Properties of Allicin from Garlic. Microbes Infect. 1999, 1, 125–129. [Google Scholar] [CrossRef]
  96. Feng, J.; Shi, W.; Miklossy, J.; Tauxe, G.M.; McMeniman, C.J.; Zhang, Y. Identification of Essential Oils with Strong Activity against Stationary Phase Borrelia burgdorferi. Antibiotics 2018, 7, 89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Salama, A.A.; AbouLaila, M.; Terkawi, M.A.; Mousa, A.; El-Sify, A.; Allaam, M.; Zaghawa, A.; Yokoyama, N.; Igarashi, I. Inhibitory effect of allicin on the growth of Babesia and Theileria equi parasites. Parasitol. Res. 2014, 113, 275–283. [Google Scholar] [CrossRef] [PubMed]
  98. Deng, S. Preliminary study on the prevention and treatment of leptospirosis with traditional Chinese medicine. Liaoning J. Tradit. Chin. Med. 1985, 9, 15–17. [Google Scholar]
  99. World Health Organization. WHO Monograph on Good Agricultural and Collection Practices (GACP) for Artemisia annua L.; World Health Organization: Geneva, Switzerland, 2006. [Google Scholar]
  100. Liu, W.; Liu, Y. Youyou Tu: Significance of winning the 2015 Nobel Prize in Physiology or Medicine. Cardiovasc. Diagn. Ther. 2016, 6, 1–2. [Google Scholar]
  101. Al-Khayri, J.M.; Sudheer, W.N.; Lakshmaiah, V.V.; Mukherjee, E.; Nizam, A.; Thiruvengadam, M.; Nagella, P.; Alessa, F.M.; Al-Mssallem, M.Q.; Rezk, A.A.; et al. Biotechnological Approaches for Production of Artemisinin, an Anti-Malarial Drug from Artemisia annua L. Molecules 2022, 27, 3040. [Google Scholar] [CrossRef]
  102. Olliaro, P.L.; Haynes, R.K.; Meunier, B.; Yuthavong, Y. Possible modes of action of the artemisinin-type compounds. Trends Parasitol. 2001, 17, 122–126. [Google Scholar] [CrossRef] [PubMed]
  103. Feng, J.; Shi, W.; Zhang, S.; Sullivan, D.; Auwaerter, P.G.; Zhang, Y. A drug combination screen identifies drugs active against amoxicillin-induced round bodies of in vitro Borrelia burgdorferi persisters from an FDA drug library. Front. Microbiol. 2016, 7, 743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Feng, J.; Weitner, M.; Shi, W.; Zhang, S.; Sullivan, D.; Zhang, Y. Identification of additional anti-persister activity against Borrelia burgdorferi from an FDA drug library. Antibiotics 2015, 4, 397–410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Krebs, S.; Omer, T.N.; Omer, B. Wormwood (Artemisia absinthium) suppresses tumor necrosis factor alpha and accelerates healing in patients with Crohn’s disease—A controlled clinical trial. Phytomedicine 2010, 17, 305–309. [Google Scholar] [CrossRef]
  106. Oryan, A. Plant-derived compounds in the treatment of leishmaniasis. Iran. J. Vet. Res. 2015, 16, 1–19. [Google Scholar]
  107. Tomosaka, H.; Chin, Y.W.; Salim, A.A.; Keller, W.J.; Chai, H.; Kinghorn, A.D. Antioxidant and cytoprotective compounds from Berberis vulgaris (Barberry). Phytother. Res. 2008, 22, 979–981. [Google Scholar] [CrossRef]
  108. Mokhber-Dezfuli, N.; Saeidnia, S.; Gohari, A.R.; Kurepaz-Mahmoodabadi, M. Phytochemistry and pharmacology of berberis species. Pharmacogn. Rev. 2014, 8, 8–15. [Google Scholar]
  109. Mahmoudvand, H.; Sharififar, F.; Sharifi, I.; Ezatpour, B.; Fasihi Harandi, M.; Makki, M.S.; Zia-Ali, N.; Jahanbakhsh, S. In vitro inhibitory effect of Berberis vulgaris (Berberidaceae) and its main component, berberine against different Leishmania species. Iran. J. Parasitol. 2014, 9, 28–36. [Google Scholar]
  110. Subeki Matsuura, H.; Takahashi, K.; Yamasaki, M.; Yamato, O.; Maede, Y.; Katakura, K.; Suzuki, M.; Trimurningsih Chairul Yoshihara, T. Antibabesial activity of protoberberine alkaloids and 20-hydroxyecdysone from Arcangelisia flava against Babesia gibsonii in culture. J. Vet. Med. Sci. 2005, 67, 223–227. [Google Scholar] [CrossRef] [Green Version]
  111. Batiha, G.E.; Magdy Beshbishy, A.; Adeyemi, O.S.; Nadwa, E.H.; Rashwan, E.K.M.; Alkazmi, L.M.; Elkelish, A.A.; Igarashi, I. Phytochemical Screening and Antiprotozoal Effects of the Methanolic Berberis vulgaris and Acetonic Rhus coriaria Extracts. Molecules 2020, 25, 550. [Google Scholar] [CrossRef] [Green Version]
  112. Elkhateeb, A.; Yamada, K.; Takahashi, K.; Matsuura, H.; Yamasaki, M.; Maede, Y.; Katakura, K.; Nabeta, K. Anti-Babesial Compounds from Berberis vulgaris. Nat. Prod. Commun. 2007, 2, 174–176. [Google Scholar] [CrossRef] [Green Version]
  113. Vasconcelos, N.; Croda, J.; Simionatto, S. Antibacterial Mechanisms of Cinnamon and its Constituents: A Review. Microb. Pathogen. 2018, 120, 198–203. [Google Scholar] [CrossRef] [PubMed]
  114. Chinou, I.; Demetzos, C.; Harvala, C.; Roussakis, C.; Verbist, J.F. Cytotoxic and antibacterial labdane-type diterpenes from the aerial parts of Cistus incanus subsp. creticus. Planta Med. 1994, 60, 34–36. [Google Scholar] [CrossRef]
  115. Yesilada, E.; Gürbüz, I.; Shibata, H. Screening of Turkish antiulcerogenic folk remedies for anti-Heliobacter pylori activity. J. Ethnopharmacol. 1999, 66, 289–293. [Google Scholar] [CrossRef] [PubMed]
  116. Demetzos, C.; Katerinopoulos, H.; Kouvarakis, A.; Stratigakis, N.; Loukis, A.; Ekonomakis, C.; Spiliotis, V.; Tsaknis, J. Composition and antimicrobial activity of the essential oil of Cistus creticus subsp. eriocephalus. Planta Med. 1997, 63, 477–479. [Google Scholar] [CrossRef]
  117. Demetzos, C.; Mitaku, S.; Couladis, M.; Harvala, C.; Kokkinopoulos, D. Natural metabolites of ent-13-epi-manoyl oxide and other cytotoxic diterpenes from the resin ladano of Cistus creticus. Planta Med. 1994, 60, 590–591. [Google Scholar] [CrossRef]
  118. Ehrhardt, C.; Hrincius, E.R.; Korte, V.; Mazur, I.; Droebner, K.; Poetter, A.; Dreschers, S.; Schmolke, M.; Planz, O.; Ludwig, S. A polyphenol rich plant extract, CYSTUS052, exerts anti influenza virus activity in cell culture without toxic side effects or the tendency to induce viral resistance. Antivir. Res 2007, 76, 38–47. [Google Scholar] [CrossRef]
  119. Güvenc, A.; Yildiz, S.; Ozkan, A.M.; Erdurak, C.S.; Coskun, M.; Yilmaz, G.; Okuyama, T.; Okada, Y. Antimicrobiological studies on Turkish Cistus species. Pharm. Biol. 2005, 43, 178–183. [Google Scholar] [CrossRef] [Green Version]
  120. Bouamama, H.; Villard, J.; Benharref, A.; Jana, M. Antibacterial and antifungal activities of Cistus incanus and C. monspeliensis leaf extracts. Therapie 1999, 54, 731–733. [Google Scholar]
  121. Bouamama, H.; Noel, T.; Villard, J.; Benharref, A.; Jana, M. Antimicrobial activities of the leaf extracts of two Moroccan Cistus L. species. J. Ethnopharmacol. 2006, 104, 104–107. [Google Scholar] [CrossRef]
  122. Petereit, F.; Kolodziej, H.; Nahrstedt, A. Flavan-3-ols and Proanthocyanidins from Cistus incanus. Phytochemistry 1991, 30, 981–985. [Google Scholar] [CrossRef]
  123. Hutschenreuther, A.; Birkemeyer, C.; Grotzinger, K.; Straubinger, R.K.; Rauwald, H.W. Growth inhibiting activity of volatile oil from Cistus creticus L. against Borrelia burgdorferi s.s. in vitro. Pharmazie 2010, 65, 290–295. [Google Scholar] [PubMed]
  124. Grellier, P.; Ramiaramanana, L.; Millerioux, V.; Deharo, E.; Schrével, J.; Frappier, F.; Trigalo, F.; Bodo, B.; Pousset, J.L. Antimalarial activity of cryptolepine and isocryptolepine, alkaloids isolated from Cryptolepis sanguinolenta. Phytother. Res. 1996, 10, 317–321. [Google Scholar] [CrossRef]
  125. Tona, L.; Kambu, K.; Ngimbi, N.; Cimanga, K.; Vlietinck, A.J. Antiamoebic and phytochemical screening of some Congolese medicinal plants. J. Ethnopharmacol. 1998, 61, 57–65. [Google Scholar] [CrossRef] [PubMed]
  126. Mills-Robertson, F.C.; Tay, S.C.; Duker-Eshun, G.; Walana, W.; Badu, K. In vitro antimicrobial activity of ethanolic fractions of Cryptolepis sanguinolenta. Ann. Clin. Microbiol. Antimicrob. 2012, 11, 16. [Google Scholar] [CrossRef] [Green Version]
  127. Ansah, C.; Mensah, K.B. A review of the anticancer potential of the antimalarial herbal Cryptolepis sanguinolenta and its major alkaloid cryptolepine. Ghana Med. J. 2013, 47, 137–147. [Google Scholar]
  128. Hanprasertpong, N.; Teekachunhatean, S.; Chaiwongsa, R.; Ongchai, S.; Kunanusorn, P.; Sangdee, C.; Panthong, A.; Bunteang, S.; Nathasaen, N.; Reutrakul, V. Analgesic, anti-inflammatory, and chondroprotective activities of Cryptolepis buchanani extract: In vitro and in vivo studies. BioMed. Res. Int. 2014, 2014, 978582. [Google Scholar] [CrossRef] [Green Version]
  129. Osafo, N.; Mensah, K.B.; Yeboah, O.K. Phytochemical and Pharmacological Review of Cryptolepis sanguinolenta (Lindl.) Schlechter. Adv. Pharmacol. Sci. 2017, 2017, 3026370. [Google Scholar] [CrossRef] [Green Version]
  130. Ameyaw, E.O.; Asmah, K.B.; Biney, R.P.; Henneh, I.T.; Owusu-Agyei, P.; Prah, J.; Forkuo, A.D. Isobolographic analysis of co-administration of two plant-derived antiplasmodial drug candidates, cryptolepine and xylopic acid, in Plasmodium berghei. Malar. J. 2018, 7, 153. [Google Scholar] [CrossRef]
  131. Bugyei, K.A.; Boye, G.L.; Addy, M.E. Clinical efficacy of a tea-bag formulation of Cryptolepis sanguinolenta root in the treatment of acute uncomplicated falciparum malaria. Ghana Med. J. 2010, 44, 3–9. [Google Scholar] [CrossRef] [Green Version]
  132. Tempesta, M.S. The clinical efficacy of Cryptolepis sanguinolenta in the treatment of malaria. Ghana Med. J. 2010, 44, 1–2. [Google Scholar] [PubMed]
  133. Ma, X.; Leone, J.; Schweig, S.; Zhang, Y. Botanical Medicines With Activity Against Stationary Phase Bartonella henselae. Infect. Microbes Dis. 2021, 3, 158–167. [Google Scholar] [CrossRef]
  134. Ajayi, A.F.; Akhigbe, R.E. Antifertility activity of Cryptolepis sanguinolenta leaf ethanolic extract in male rats. J. Hum. Reprod. Sci. 2012, 5, 43–47. [Google Scholar] [PubMed]
  135. Mensah, K.B.; Benneh, C.; Forkuo, A.D.; Ansah, C. Cryptolepine, the Main Alkaloid of the Antimalarial Cryptolepis sanguinolenta (Lindl.) Schlechter, Induces Malformations in Zebrafish Embryos. Biochem. Res. Int. 2019, 2019, 7076986. [Google Scholar] [CrossRef] [Green Version]
  136. Liebold, T.; Straubinger, R.; Rauwald, H. Growth inhibiting activity of lipophilic extracts from Dipsacus sylvestris huds. Roots against Borrelia burgdorferi s. s. in vitro. Pharmazie 2011, 66, 628–630. [Google Scholar] [PubMed]
  137. Goc, A.; Rath, M. The anti-borreliae efficacy of phytochemicals and micronutrients: An update. Ther. Adv. Infect. Dis. 2016, 3, 75–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Batiha, G.E.; Alkazmi, L.M.; Wasef, L.G.; Beshbishy, A.M.; Nadwa, E.H.; Rashwan, E.K. Syzygium aromaticum L. (Myrtaceae): Traditional Uses, Bioactive Chemical Constituents, Pharmacological and Toxicological Activities. Biomolecules 2020, 10, 202. [Google Scholar] [CrossRef] [Green Version]
  139. Batiha, G.E.; Beshbishy, A.M.; Tayebwa, D.S.; Shaheen, H.M.; Yokoyama, N.; Igarashi, I. Inhibitory effects of Syzygium aromaticum and Camellia sinensis methanolic extracts on the growth of Babesia and Theileria parasites. Ticks Tick. Borne Dis. 2019, 10, 949–958. [Google Scholar] [CrossRef]
  140. Bi, D.; Zhao, Y.; Jiang, R.; Wang, Y.; Tian, Y.; Chen, X.; Bai, S.; She, G. Phytochemistry, bioactivity and potential impact on health of Juglans: The original plant of walnut. Nat. Prod. Commun. 2016, 11, 869–880. [Google Scholar] [CrossRef] [Green Version]
  141. Ahmad, T.; Suzuki, Y.J. Juglone in oxidative stress and cell signaling. Antioxidants 2019, 8, 91. [Google Scholar] [CrossRef] [Green Version]
  142. Therapeutic Research Center. Natural Medicines Monograph: Black Walnut. 2019. Available online: https://naturalmedicines.therapeuticresearch.com/ (accessed on 4 March 2019).
  143. Bonamonte, D.; Foti, C.; Angelini, G. Hyperpigmentation and contact dermatitis due to Juglans regia. Contact Derm. 2001, 44, 102–103. [Google Scholar] [CrossRef]
  144. Neri, I.; Bianchi, F.; Giacomini, F.; Patrizi, A. Acute irritant contact dermatitis due to Juglans regia. Contact Derm. 2006, 55, 62–63. [Google Scholar] [CrossRef] [PubMed]
  145. Siegel, J.M. Dermatitis due to black walnut juice. AMA Arch. Derm. Syphilol. 1954, 70, 511–513. [Google Scholar] [CrossRef] [PubMed]
  146. Barker, L.A.; Bakkum, B.W.; Chapman, C. The Clinical Use of Monolaurin as a Dietary Supplement: A Review of the Literature. J. Chiropr. Med. 2019, 18, 305–310. [Google Scholar] [CrossRef]
  147. Batovska, D.I.; Todorova, I.T.; Tsvetkova, I.V.; Najdenski, H. MAntibacterial study of the medium chain fatty acids and their 1-monoglycerides: Individual effects and synergistic relationships. Pol. J. Microbiol. 2009, 58, 43–47. [Google Scholar]
  148. El-Sayed, S.A.E.; Rizk, M.A.; Yokoyama, N.; Igarashi, I. Evaluation of the in vitro and in vivo inhibitory effect of thymoquinone on piroplasm parasites. Parasit Vectors. 2019, 12, 37. [Google Scholar] [CrossRef]
  149. McCormick Science Institute. Resources: Oregano. Available online: https://www.mccormickscienceinstitute.com/resources/culinary-spices/herbs-spices/oregano#:~:text=Oregano%20is%20the%20dried%20leaves,in%20high%20altitude%20Mediterranean%20climates.> (accessed on 14 January 2023).
  150. Melo, F.H.C.; Moura, B.A.; de Sousa, D.P.; de Vasconcelos, S.M.M.; Macedo, D.S.; Fonteles, M.M.d.F.; Viana, G.S.d.B.; de Sousa, F.C.F. Antidepressant-like effect of carvacrol (5-isopropyl-2- methylphenol) in mice: Involvement of dopaminergic system. Fundam. Clin. Pharm. 2011, 25, 362–367. [Google Scholar] [CrossRef]
  151. Wu, X.; Li, Q.; Feng, Y.; Ji, Q. Antitumor research of the active ingredients from traditional Chinese medical plant Polygonum cuspidatum. Evid.-Based Complement. Altern. Med. 2018, 2018, 2313021. [Google Scholar] [CrossRef] [Green Version]
  152. Breuss, J.M.; Atanasov, A.G.; Uhrin, P. Resveratrol and its effects on the vascular system. Int. J. Mol. Sci. 2019, 20, 1523. [Google Scholar] [CrossRef] [Green Version]
  153. Kim, J.R.; Oh, D.R.; Cha, M.H.; Pyo, B.S.; Rhee, J.H.; Choy, H.E.; Oh, W.K.; Kim, Y.R. Protective effect of polygoni cuspidati radix and emodin on Vibrio vulnificus cytotoxicity and infection. J. Microbiol. 2008, 46, 737–743. [Google Scholar] [CrossRef]
  154. Pandit, S.; Kim, H.J.; Park, S.H.; Jeon, J.G. Enhancement of fluoride activity against Streptococcus mutans biofilms by a substance separated from Polygonum cuspidatum. Biofouling 2012, 28, 279–287. [Google Scholar] [CrossRef]
  155. Shan, B.; Cai, Y.Z.; Brooks, J.D.; Corke, H. Antibacterial properties of Polygonum cuspidatum roots and their major bioactive constituents. Food Chem. 2008, 109, 530–537. [Google Scholar] [CrossRef]
  156. Ghanim, H.; Sia, C.L.; Abuaysheh, S.; Korzeniewski, K.; Patnaik, P.; Marumganti, A.; Chaudhuri, A.; Dandona, P. An antiinflammatory and reactive oxygen species suppressive effects of an extract of Polygonum cuspidatum containing resveratrol. J. Clin. Endocrinol. Metab. 2010, 95, E1–E8. [Google Scholar] [CrossRef] [Green Version]
  157. la Porte, C.; Voduc, N.; Zhang, G.; Seguin, I.; Tardiff, D.; Singhal, N.; Cameron, D.W. Steady-state pharmacokinetics and tolerability of trans-resveratrol 2000mg twice daily with food, Quercetin and Alcohol (Ethanol) in healthy human subjects. Clin. Pharmacokinet. 2010, 49, 449–454. [Google Scholar] [CrossRef] [PubMed]
  158. Alsamri, H.; Athamneh, K.; Pintus, G.; Eid, A.H.; Iratni, R. Pharmacological and Antioxidant Activities of Rhus coriaria L. (Sumac). Antioxidants 2021, 10, 73. [Google Scholar] [CrossRef] [PubMed]
  159. Deguchi, Y.; Ito, M. Rosmarinic acid in Perilla frutescens and perilla herb analyzed by HPLC. J. Nat. Med. 2020, 74, 341–352. [Google Scholar] [CrossRef]
  160. Goc, A.; Niedzwiecki, A.; Rath, M. Reciprocal cooperation of phytochemicals and micronutrients against typical and atypical forms of Borrelia sp. J. Appl. Microbiol. 2017, 123, 637–650. [Google Scholar] [CrossRef]
  161. Luo, J.; Dong, B.; Wang, K.; Cai, S.; Liu, T.; Cheng, X.; Lei, D.; Chen, Y.; Li, Y.; Kong, J.; et al. Baicalin inhibits biofilm formation, attenuates the quorum sensing-controlled virulence and enhances Pseudomonas aeruginosa clearance in a mouse peritoneal implant infection model. PLoS ONE 2017, 12, e0176883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Fujita, M.; Shiota, S.; Kuroda, T.; Hatano, T.; Yoshida, T.; Mizushima, T.; Tsuchiya, T. Remarkable synergies between baicalein and tetracycline, and baicalein and b-lactams against methicillin-resistant Staphylococcus aureus. Microbiol. Immunol. 2005, 49, 391–396. [Google Scholar] [CrossRef]
  163. Cai, W.; Fu, Y.; Zhang, W.; Chen, X.; Zhao, J.; Song, W.; Li, Y.; Huang, Y.; Wu, Z.; Sun, R.; et al. Synergistic effects of baicalein with cefotaxime against Klebsiella pneumoniae through inhibiting CTX-M-1 gene expression. BMC Microbiol. 2016, 16, 181. [Google Scholar] [CrossRef] [Green Version]
  164. Wang, J.; Qiao, M.; Zhou, Y.; Du, H.; Bai, J.; Yuan, W.; Liu, J.; Wang, D.; Hu, Y.; Wu, Y. In vitro synergistic effect of baicalin with azithromycin against Staphylococcus saprophyticus isolated from francolins with ophthalmia. Poult. Sci. 2019, 98, 373–380. [Google Scholar] [CrossRef]
  165. Gol’Dberg, V.E.; Ryzhakov, V.M.; Matiash, M.G.; Stepovaia, E.A.; Boldyshev, D.A.; Litvinenko, V.I.; Dygaĭ, A.M. Dry extract of Scutellaria baicalensis as a hemostimulant in antineoplastic chemotherapy in patents with lung cancer. Eksp. Klin. Farmakol. 1997, 60, 28–30. [Google Scholar] [PubMed]
  166. Smol’Ianinov, E.S.; Gol’Dberg, V.E.; Matiash, M.G.; Ryzhakov, V.M.; Boldyshev, D.A.; Litvinenko, V.I.; Dygaĭ, A.M. Effect of Scutellaria baicalensis extract on the immunologic status of patients with lung cancer receiving antineoplastic chemotherapy. Eksp. Klin. Farmakol. 1997, 60, 49–51. [Google Scholar]
  167. Zhou, H.C.; Wang, H.; Shi, K.; Li, J.M.; Zong, Y.; Du, R. Hepatoprotective Effect of Baicalein Against Acetaminophen-Induced Acute Liver Injury in Mice. Molecules 2018, 24, 131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Hui, K.M.; Wang, X.H.; Xue, H. Interaction of flavones from the roots of Scutellaria baicalensis with the benzodiazepine site. Planta Med. 2000, 66, 91–93. [Google Scholar] [CrossRef]
  169. Theophilus, P.A.; Victoria, M.J.; Socarras, K.M.; Filush, K.R.; Gupta, K.; Luecke, D.F.; Sapi, E. Effectiveness of Stevia rebaudiana Whole Leaf Extract Against the Various Morphological Forms of Borrelia Burgdorferi in Vitro. Eur. J. Microbiol. Immunol. 2015, 5, 268–280. [Google Scholar] [CrossRef] [Green Version]
  170. Anton, S.D.; Martin, C.K.; Han, H.; Coulon, S.; Cefalu, W.T.; Geiselman, P.; Williamson, D.A. Effects of stevia, aspartame, and sucrose on food intake, satiety, and postprandial glucose and insulin levels. Appetite 2010, 55, 37–43. [Google Scholar] [CrossRef] [Green Version]
  171. Carakostas, M.C.; Curry, L.L.; Boileau, A.C.; Brusick, D.J. Overview: The history, technical function and safety of rebaudioside A, a naturally occurring steviol glycoside, for use in food and beverages. Food Chem. Toxicol. 2008, 46 (Suppl. 7), S1–S10. [Google Scholar] [CrossRef] [PubMed]
  172. Herrera, D.R.; Durand-Ramirez, J.E.; Falcao, A.; Silva, E.J.; Santos, E.B.; Gomes, B.P. Antimicrobial activity and substantivity of Uncaria tomentosa in infected root canal dentin. Braz. Oral Res. 2016, 30, e61. [Google Scholar] [CrossRef] [Green Version]
  173. Sheng, Y.; Li, L.; Holmgren, K.; Pero, R.W. DNA repair enhancement of aqueous extracts of Uncaria tomentosa in a human volunteer study. Phytomedicine 2001, 8, 275–282. [Google Scholar] [CrossRef] [Green Version]
  174. Piscoya, J.; Rodriguez, Z.; Bustamante, S.A.; Okuhama, N.N.; Miller, M.J.; Sandoval, M. Efficacy and safety of freeze-dried cat’s claw in osteoarthritis of the knee: Mechanisms of action of the species Uncaria guianensis. Inflamm. Res. 2001, 50, 442–448. [Google Scholar] [CrossRef]
  175. Mur, E.; Hartig, F.; Eibl, G.; Schirmer, M. Randomized double-blind trial of an extract from the pentacyclic alkaloid-chemotype of Uncaria tomentosa for the treatment of rheumatoid arthritis. J. Rheumatol. 2002, 29, 678–681. [Google Scholar]
  176. Salazar, E.L.; Jayme, V. Depletion of specific binding sites for estrogen receptor by Uncaria tomentosa. Proc. West. Pharmacol. Soc. 1998, 41, 123–124. [Google Scholar] [PubMed]
  177. Neto, J.N.; Cavalcante, F.L.L.P.; Carvalho, R.A.F.; Rodrigues, T.G.P.D.M.; Xavier, M.S.; Furtado, P.G.R.; Schor, E. Contraceptive effect of Uncaria tomentosa (cat’s claw) in rats with experimental endometriosis. Acta Cirugica Bras. 2011, 26 (Suppl. S2), 15–19. [Google Scholar] [CrossRef] [Green Version]
  178. Goc, A.; Niedzwiecki, A.; Rath, M. Cooperation of Doxycycline with Phytochemicals and Micronutrients Against Active and Persistent Forms of Borrelia sp. Int. J. Biol. Sci. 2016, 12, 1093–1103. [Google Scholar] [CrossRef] [Green Version]
  179. Donta, S.T. Issues in the diagnosis and treatment of Lyme disease. Open Neurol. J. 2012, 6, 140–145. [Google Scholar] [CrossRef] [Green Version]
  180. Rizk, M.A.; El-Sayed, S.A.E.; Igarashi, I. Ascorbic acid co-administration with a low dose of diminazene aceturate inhibits the in vitro growth of Theileria equi, and the in vivo growth of Babesia microti. Parasitol. Int. 2022, 90, 102596. [Google Scholar] [CrossRef] [PubMed]
  181. Cantorna, M.T. Vitamin D, multiple sclerosis and inflammatory bowel disease. Arch. Biochem. Biophys. 2012, 523, 103–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Cantorna, M.T.; Hayes, C.E.; DeLuca, H.F. 1,25-Dihydroxycholecalciferol inhibits the progression of arthritis in murine models of human arthritis. J. Nutr. 1998, 128, 68–72. [Google Scholar] [CrossRef] [Green Version]
  183. Goc, A.; Gehring, G.; Baltin, H.; Niedzwiecki, A.; Rath, M. Specific composition of polyphenolic compounds with fatty acids as an approach in helping to reduce spirochete burden in Lyme disease: In vivo and human observational study. Ther. Adv. Chronic Dis. 2020, 11, 2040622320922005. [Google Scholar] [CrossRef] [PubMed]
  184. Pushparajah Mak, R.S.; Liebelt, E.L. Methylene Blue: An Antidote for Methemoglobinemia and Beyond. Pediatr. Emerg. Care 2021, 37, 474–477. [Google Scholar] [CrossRef]
  185. Zoungrana, A.; Coulibaly, B.; Sié, A.; Walter-Sack, I.; Mockenhaupt, F.P.; Kouyaté, B.; Schirmer, R.H.; Klose, C.; Mansmann, U.; Meissner, P.; et al. Safety and efficacy of methylene blue combined with artesunate or amodiaquine for uncomplicated falciparum malaria: A randomized controlled trial from Burkina Faso. PLoS ONE 2008, 3, e1630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Kwok, E.S.; Howes, D. Use of methylene blue in sepsis: A systematic review. J. Intensive Care Med. 2006, 21, 359–363. [Google Scholar] [CrossRef] [PubMed]
  187. Hillary, S.L.; Guillermet, S.; Brown, N.J.; Balasubramanian, S.P. Use of methylene blue and near-infrared fluorescence in thyroid and parathyroid surgery. Langenbecks Arch. Surg. 2018, 403, 111–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Paul, A.S.; Moreira, C.K.; Elsworth, B.; Allred, D.R.; Duraisingh, M.T. Extensive Shared Chemosensitivity between Malaria and Babesiosis Blood-Stage Parasites. Antimicrob. Agents Chemother. 2016, 60, 5059–5063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Ord, R.L.; Lobo, C.A. Human Babesiosis: Pathogens, Prevalence, Diagnosis, and Treatment. Curr. Clin. Microbiol. Rep. 2015, 2, 173–181. [Google Scholar] [CrossRef] [Green Version]
  190. Carvalho, L.J.M.; Tuvshintulga, B.; Nugraha, A.B.; Sivakumar, T.; Yokoyama, N. Activities of artesunate-based combinations and tafenoquine against Babesia bovis in vitro and Babesia microti in vivo. Parasits Vectors 2020, 13, 362. [Google Scholar] [CrossRef]
  191. Galkin, A.; Kulakova, L.; Lim, K.; Chen, C.Z.; Zheng, W.; Turko, I.V.; Herzberg, O. Structural basis for inactivation of Giardia lamblia carbamate kinase by disulfiram. J. Biol. Chem. 2014, 289, 10502–10509. [Google Scholar] [CrossRef] [Green Version]
  192. Hao, W.; Qiao, D.; Han, Y.; Du, N.; Li, X.; Fan, Y.; Ge, X.; Zhang, H. Identification of disulfiram as a potential antifungal drug by screening small molecular libraries. J. Infect. Chemother. 2021, 27, 696–701. [Google Scholar] [CrossRef]
  193. Long, T.E. Repurposing Thiram and Disulfiram as Antibacterial Agents for Multidrug-Resistant Staphylococcus aureus Infections. Antimicrob. Agents Chemother. 2017, 61, e00898-17. [Google Scholar] [CrossRef] [Green Version]
  194. Horita, Y.; Takii, T.; Yagi, T.; Ogawa, K.; Fujiwara, N.; Inagaki, E.; Kremer, L.; Sato, Y.; Kuroishi, R.; Lee, Y.; et al. Antitubercular activity of disulfiram, an antialcoholism drug, against multidrug-and extensively drug-resistant Mycobacterium tuberculosis isolates. Antimicrob. Agents Chemother. 2012, 56, 4140–4145. [Google Scholar] [CrossRef] [Green Version]
  195. Scheibel, L.W.; Adler, A.; Trager, W. Tetraethylthiuram disulfide (Antabuse) inhibits the human malaria parasite Plasmodium falciparum. Proc. Natl. Acad. Sci. USA 1979, 76, 5303–5307. [Google Scholar] [CrossRef] [PubMed]
  196. Potula, H.S.K.; Shahryari, J.; Inayathullah, M.; Malkovskiy, A.V.; Kim, K.M.; Rajadas, J. Repurposing Disulfiram (Tetraethylthiuram Disulfide) as a Potential Drug Candidate against Borrelia burgdorferi In Vitro and In Vivo. Antibiotics 2020, 9, 633. [Google Scholar] [CrossRef] [PubMed]
  197. Pothineni, V.; Wagh, D.; Babar, M.M.; Inayathullah, M.; Solow-Cordero, D.; Kim, K.-M.; Samineni, A.; Parekh, M.B.; Tayebi, L.; Rajadas, J. Identification of new drug candidates against Borrelia burgdorferi using high-throughput screening. Drug. Des. Devel. Ther. 2016, 10, 1307. [Google Scholar] [CrossRef] [Green Version]
  198. Liegner, K.B. Disulfiram (Tetraethylthiuram Disulfide) in the Treatment of Lyme Disease and Babesiosis: Report of Experience in Three Cases. Antibiotics 2019, 8, 72. [Google Scholar] [CrossRef] [Green Version]
  199. Gao, J.; Gong, Z.; Montesano, D.; Glazer, E.; Liegner, K. “Repurposing” Disulfiram in the Treatment of Lyme Disease and Babesiosis: Retrospective Review of First 3 Years’ Experience in One Medical Practice. Antibiotics 2020, 9, 868. [Google Scholar] [CrossRef] [PubMed]
  200. Di Lorenzo, C.; Ceschi, A.; Kupferschmidt, H.; Lüde, S.; De Souza Nascimento, E.; Dos Santos, A.; Colombo, F.; Frigerio, G.; Nørby, K.; Plumb, J.; et al. Adverse effects of plant food supplements and botanical preparations: A systematic review with critical evaluation of causality. Br. J. Clin. Pharmacol. 2015, 79, 578–592. [Google Scholar] [CrossRef] [Green Version]
  201. Perrillo, R.P.; Burton, J.R., Jr.; Westbrook, L.M. Herbal hepatitis due to use of alternative medicines for Lyme disease. Bayl. Univ. Med. Cent. Proc. 2021, 35, 104–105. [Google Scholar] [CrossRef]
Table 1. Research by Goc and colleagues [67]; analyzed organic oil for activity against 3 forms of Borrelia spp.: active, stationary, and biofilm persister forms.
Table 1. Research by Goc and colleagues [67]; analyzed organic oil for activity against 3 forms of Borrelia spp.: active, stationary, and biofilm persister forms.
Organic OilsActive IngredientActiveStationaryBiofilm
Bay leaf oil (Pimenta racemosa)EugenolXXX
Birch (sweet) oil (Betula lenta)Methyl salicylate XXX
Cassia oil (Cinnamomum cassia) CinnamaldehydeXXX
Chamomile oil German (Matricaria chamomilla)Chamazulene XXX
Thyme oil (Thymus vulgaris)ThymolXXX
Table 2. Summarizing various combinations of agents with synergistic or additive impact on spirochetal, stationary and biofilm forms against the two strains of Borrelia studied.
Table 2. Summarizing various combinations of agents with synergistic or additive impact on spirochetal, stationary and biofilm forms against the two strains of Borrelia studied.
Synergistic or Additive CombinationsSpirocheteStationaryBiofilm
Baicalein with luteolin XXX
Monolaurin with cis-2-decenoic acid X X
Baicalein and rosmarinic acid X
Luteolin and rosmarinic acid X
Baicalein and iodine X
Luteolin and iodine X
Baicalein with cis-2-decenoic acid X
Luteolin with cis-2-decenoic acid X
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Shor, S.M.; Schweig, S.K. The Use of Natural Bioactive Nutraceuticals in the Management of Tick-Borne Illnesses. Microorganisms 2023, 11, 1759. https://doi.org/10.3390/microorganisms11071759

AMA Style

Shor SM, Schweig SK. The Use of Natural Bioactive Nutraceuticals in the Management of Tick-Borne Illnesses. Microorganisms. 2023; 11(7):1759. https://doi.org/10.3390/microorganisms11071759

Chicago/Turabian Style

Shor, Samuel M., and Sunjya K. Schweig. 2023. "The Use of Natural Bioactive Nutraceuticals in the Management of Tick-Borne Illnesses" Microorganisms 11, no. 7: 1759. https://doi.org/10.3390/microorganisms11071759

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop