Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Shashikant Srivastava is active.

Publication


Featured researches published by Shashikant Srivastava.


The Journal of Infectious Diseases | 2011

Multidrug-Resistant Tuberculosis Not Due to Noncompliance but to Between-Patient Pharmacokinetic Variability

Shashikant Srivastava; Jotam G. Pasipanodya; Claudia Meek; Richard Leff; Tawanda Gumbo

BACKGROUND It is believed that nonadherence is the proximate cause of multidrug-resistant tuberculosis (MDR-tuberculosis) emergence. The level of nonadherence associated with emergence of MDR-tuberculosis is unknown. Performance of a randomized controlled trial in which some patients are randomized to nonadherence would be unethical; therefore, other study designs should be utilized. METHODS We performed hollow fiber studies for both bactericidal and sterilizing effect, with inoculum spiked with 0.5% rifampin- and isoniazid-resistant isogenic strains in some experiments. Standard therapy was administered daily for 28-56 days, with extents of nonadherence varying between 0% and 100%. Sizes of drug-resistant populations were compared using analysis of variance. We also explored the effect of pharmacokinetic variability on MDR-tuberculosis emergence using computer-aided clinical trial simulations of 10 000 Cape Town, South Africa, tuberculosis patients. RESULTS Therapy failure was only encountered at extents of nonadherence ≥60%. Surprisingly, isoniazid- and rifampin-resistant populations did not achieve ≥1% proportion in any experiment and did not achieve a higher proportion with nonadherence. However, clinical trial simulations demonstrated that approximately 1% of tuberculosis patients with perfect adherence would still develop MDR-tuberculosis due to pharmacokinetic variability alone. CONCLUSIONS These data, based on a preclinical model, demonstrate that nonadherence alone is not a sufficient condition for MDR-tuberculosis emergence.


Clinical Infectious Diseases | 2012

Meta-Analysis of Clinical Studies Supports the Pharmacokinetic Variability Hypothesis for Acquired Drug Resistance and Failure of Antituberculosis Therapy

Jotam G. Pasipanodya; Shashikant Srivastava; Tawanda Gumbo

BACKGROUND Using hollow-fiber tuberculosis studies, we recently demonstrated that nonadherence is not a significant factor for ADR and that therapy failure only occurs after a large proportion of doses are missed. Computer-aided clinical trial simulations have suggested that isoniazid and rifampin pharmacokinetic variability best explained poor outcomes. We were interested in determining whether isoniazid pharmacokinetic variability was associated with either microbiological failure or ADR in the clinic. METHODS Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were followed. Prospective, randomized, controlled clinical trials that reported isoniazid acetylation status and microbiological outcomes were selected. The main effects examined were microbiological sputum conversion, ADR, and relapse. Effect size was expressed as pooled risk ratios (RRs) comparing rapid with slow acetylators. RESULTS Thirteen randomized studies with 1631 rapid acetylators and 1751 slow acetylators met inclusion and exclusion criteria. Rapid acetylators were more likely than slow acetylators to have microbiological failure (RR, 2.0; 95% confidence interval [CI], 1.5-2.7), ADR (RR, 2.0; CI, 1.1-3.4), and relapse (RR, 1.3; CI, .9-2.0). Higher failure rates were encountered even in drug regimens comprising >3 antibiotics. No publication bias or small-study effects were observed for the outcomes evaluated. CONCLUSIONS Pharmacokinetic variability to a single drug in the regimen is significantly associated with failure of therapy and ADR in patients. This suggests that individualized dosing for tuberculosis may be more effective than standardized dosing, which is prescribed in directly observed therapy programs.


Antimicrobial Agents and Chemotherapy | 2012

The Antibiotic-Resistance Arrow of Time: Efflux Pump Induction is a General First Step in the Evolution of Mycobacterial Drug-Resistance

Aurelia Schmalstieg; Shashikant Srivastava; Serkan Belkaya; Devyani Deshpande; Claudia Meek; Richard Leff; Nicolai S. C. van Oers; Tawanda Gumbo

ABSTRACT We hypothesize that low-level efflux pump expression is the first step in the development of high-level drug resistance in mycobacteria. We performed 28-day azithromycin dose-effect and dose-scheduling studies in our hollow-fiber model of disseminated Mycobacterium avium-M. intracellulare complex. Both microbial kill and resistance emergence were most closely linked to the within-macrophage area under the concentration-time curve (AUC)/MIC ratio. Quantitative PCR revealed that subtherapeutic azithromycin exposures over 3 days led to a 56-fold increase in expression of MAV_3306, which encodes a putative ABC transporter, and MAV_1406, which encodes a putative major facilitator superfamily pump, in M. avium. By day 7, a subpopulation of M. avium with low-level resistance was encountered and exhibited the classic inverted U curve versus AUC/MIC ratios. The resistance was abolished by an efflux pump inhibitor. While the maximal microbial kill started to decrease after day 7, a population with high-level azithromycin resistance appeared at day 28. This resistance could not be reversed by efflux pump inhibitors. Orthologs of pumps encoded by MAV_3306 and MAV_1406 were identified in Mycobacterium tuberculosis, Mycobacterium leprae, Mycobacterium marinum, Mycobacterium abscessus, and Mycobacterium ulcerans. All had highly conserved protein secondary structures. We propose that induction of several efflux pumps is the first step in a general pathway to drug resistance that eventually leads to high-level chromosomal-mutation-related resistance in mycobacteria as ordered events in an “antibiotic resistance arrow of time.”


The Journal of Infectious Diseases | 2010

Efflux-Pump—Derived Multiple Drug Resistance to Ethambutol Monotherapy in Mycobacterium tuberculosis and the Pharmacokinetics and Pharmacodynamics of Ethambutol

Shashikant Srivastava; Sandirai Musuka; Carleton Sherman; Claudia Meek; Richard Leff; Tawanda Gumbo

BACKGROUND Ethambutol is used for the treatment of tuberculosis in cases where there is isoniazid resistance. We examined the emergence of drug resistance to ethambutol monotherapy in pharmacokinetic-pharmacodynamic studies of a hollow-fiber system. METHODS Dose-effect and dose-scheduling studies were performed with ethambutol and log-phase growth Mycobacterium tuberculosis to identify exposures and schedules linked to optimal kill and resistance suppression. In one study, after 7 days of daily ethambutol, 300 mg isoniazid per day was administered to each system to determine its early bactericidal activity. RESULTS Efflux-pump blockage reduced the mutation frequency to ethambutol 64-fold. In dose-effect studies, ethambutol had a maximal early bactericidal activity of 0.22 log10 colony-forming units/mL/day, as is encountered in patients. By day 7, resistance to both ethambutol and isoniazid had increased. Previous exposure to ethambutol halted isoniazid early bactericidal activity. Daily therapy, as opposed to more intermittent therapy, was associated with the least proportion of efflux-pump-driven resistance, consistent with a time-driven effect. Microbial kill was best explained by the ratio of area under the concentration-time curve to minimum inhibitory concentration (r2 = 0.90). CONCLUSION The induction of an efflux pump that reduces the effect of multiple drugs provides an alternative pathway to sequential acquisition of mutations in the development of multiple drug resistance.


Antimicrobial Agents and Chemotherapy | 2010

Ethambutol Optimal Clinical Dose and Susceptibility Breakpoint Identification by Use of a Novel Pharmacokinetic-Pharmacodynamic Model of Disseminated Intracellular Mycobacterium avium

Devyani Deshpande; Shashikant Srivastava; Claudia Meek; Richard Leff; Tawanda Gumbo

ABSTRACT Ethambutol, together with a macrolide, is the backbone for treatment of disseminated Mycobacterium avium disease. However, at the standard dose of 15 mg/kg of body weight/day, ethambutol efficacy is limited. In addition, susceptibility breakpoints have consistently failed to predict clinical outcome. We performed dose-effect studies with extracellular M. avium as well as with bacilli within human macrophages. The maximal kill rate (Emax) for ethambutol against extracellular bacilli was 5.54 log10 CFU/ml, compared to 0.67 log10 CFU/ml for intracellular M. avium, after 7 days of exposure. Thus, extracellular assays demonstrated high efficacy. We created a hollow-fiber system model of intracellular M. avium and performed microbial pharmacokinetic-pharmacodynamic studies using pharmacokinetics similar to those of ethambutol for humans. The Emax in the systems was 0.79 log10 CFU/ml with 7 days of daily therapy, so the kill rates approximated those encountered in patients treated with ethambutol monotherapy. Ratio of peak concentration to MIC (Cmax/MIC) was linked to microbial kill rate. The Cmax/MIC ratio needed to achieve the 90% effective concentration (EC90) in serum was 1.23, with a calculated intramacrophage Cmax/MIC ratio of 13. In 10,000 patient Monte Carlo simulations, doses of 15, 50, and 75 mg/kg achieved the EC90 in 35.50%, 76.81%, and 86.12% of patients, respectively. Therefore, ethambutol doses of ≥50 mg/kg twice a week would be predicted to be better than current doses of 15 mg/kg for treatment of disseminated M. avium disease. New susceptibility breakpoints and critical concentrations of 1 to 2 mg/liter were identified for the determination of ethambutol-resistant M. avium in Middlebrook broth. Given that the modal MIC of clinical isolates is around 2 mg/liter, most isolates should be considered ethambutol resistant.


European Respiratory Journal | 2013

Therapeutic drug management: is it the future of multidrug-resistant tuberculosis treatment?

Shashikant Srivastava; Charles A. Peloquin; Giovanni Sotgiu; Giovanni Battista Migliori

Multidrug- and extensively drug-resistant (M/XDR) tuberculosis (TB) are emerging public health concerns [1, 2]. In 2011, the World Health Organization (WHO) estimated 12 million prevalent cases of TB globally, which is equivalent to 170 cases per 100 000 population, out of these an estimated 630 000 cases were affected by MDR Mycobacterium tuberculosis strains [3]. Among the newly diagnosed patients ∼3.7% were infected by MDR-TB strains, but the worrisome fact is that the prevalence of MDR-TB among new cases in some Former Soviet Union countries exceeds 30% [4, 5], XDR-TB has been identified in 84 countries and the average proportion of MDR-TB cases with an XDR-TB pattern is 9.0% [3]. Further adding to the problem are the reports of “totally drug resistant” TB [6, 7], a term currently not recognised by WHO [8, 9]. Treatment of drug resistant TB is more expensive and more toxic if compared with that prescribed for drug-susceptible TB, and currently takes up to 2 years of therapy [10]. The cost per patient to treat MDR-TB cases is incredibly high [11, 12] and, in spite of international public health efforts, the treatment outcome is not very promising [13–15]. Diel et al. [16] showed that direct treatment-related costs of MDR-TB patients can amount to €52 259 in Germany (table 1). View this table: Table 1– Direct costs of multidrug-resistant tuberculosis (MDR-TB) therapy in a European low-income country In the largest MDR-TB cohort analysed to date [13] the proportion of cases treated successfully was 62%, with 7% failing or relapsing, 9% dying and 17% defaulting; in the XDR-TB subgroup 40% achieved treatment success, 22% failed treatment or relapsed, whereas 15% died and 16% defaulted [14, 15]. In this issue of the …


Antimicrobial Agents and Chemotherapy | 2011

Pharmacokinetic Mismatch Does Not Lead to Emergence of Isoniazid- or Rifampin-Resistant Mycobacterium tuberculosis but to Better Antimicrobial Effect: a New Paradigm for Antituberculosis Drug Scheduling

Shashikant Srivastava; Carleton Sherman; Claudia Meek; Richard Leff; Tawanda Gumbo

ABSTRACT Multidrug resistant-tuberculosis is a pressing problem. One of the major mechanisms proposed to lead to the emergence of drug resistance is pharmacokinetic mismatch. Stated as a falsifiable hypothesis, the greater the pharmacokinetic mismatch between rifampin and isoniazid, the higher the isoniazid- and rifampin-resistant subpopulation sizes become with time. To test this, we performed hollow-fiber-system studies for both bactericidal and sterilizing effects in experiments of up to 42 days. We mimicked pharmacokinetics of 600-mg/day rifampin and 300-mg/day isoniazid administered to patients. Rifampin was administered first, followed by isoniazid 0, 6, 12, and 24 h later. The treatment was for drug-susceptible Mycobacterium tuberculosis in some experiments and hollow fiber systems with inoculum preseeded with isoniazid- and rifampin-resistant isogenic Mycobacterium tuberculosis strains in others. Analysis of variance revealed that the 12-h and 24-h-mismatched regimens always killed better than the matched regimens during both bactericidal and sterilizing effects (P < 0.05). This means that either the order of scheduling or the sequential administration of drugs in combination therapy may lead to significant improvement in microbial killing. Rifampin-resistant and isoniazid-resistant subpopulations were not significantly higher with increased mismatching in numerous analysis-of-variance comparisons. Thus, the pharmacokinetic mismatch hypothesis was rejected. Instead, sequential administration of anti-tuberculosis (TB) drugs (i.e., deliberate mismatch) following particular schedules suggests a new paradigm for accelerating M. tuberculosis killing. We conclude that current efforts aimed at better pharmacokinetic matching to decrease M. tuberculosis resistance emergence are likely futile and counterproductive.


Antimicrobial Agents and Chemotherapy | 2010

Moxifloxacin Pharmacokinetics/Pharmacodynamics and Optimal Dose and Susceptibility Breakpoint Identification for Treatment of Disseminated Mycobacterium avium Infection

Devyani Deshpande; Shashikant Srivastava; Claudia Meek; Richard Leff; Gerri S. Hall; Tawanda Gumbo

ABSTRACT Organisms of the Mycobacterium avium-intracellulare complex (MAC) have been demonstrated to be susceptible to moxifloxacin. However, clinical data on how to utilize moxifloxacin to treat disseminated MAC are scanty. In addition, there have been no moxifloxacin pharmacokinetic-pharmacodynamic (PK/PD) studies performed for MAC infection. We utilized an in vitro PK/PD model of intracellular MAC to study moxifloxacin PK/PD for disseminated disease. Moxifloxacin doses, based on a serum half-life of 12 h, were administered, and the 0- to 24-h area under the concentration-time curve (AUC0-24) to MIC ratios associated with 1.0 log10 CFU/ml per week kill and 90% of maximal kill (EC90) were identified. The AUC0-24/MIC ratio associated with 1.0 log10 CFU/ml kill was 17.12, and that with EC90 was 391.56 (r2 = 0.97). Next, the moxifloxacin MIC distribution in 102 clinical isolates of MAC was identified. The median MIC was 1 to 2 mg/liter. Monte Carlo simulations of 10,000 patients with disseminated MAC were performed to determine the probability that daily moxifloxacin doses of 400 and 800 mg/day would achieve or exceed 1.0 log10 CFU/ml per week kill or EC90. Doses of 400 and 800 mg/day achieved the AUC0-24/MIC ratio of 17.12 in 64% and 92% of patients, respectively. The critical concentration of moxifloxacin against MAC was identified as 0.25 mg/liter in Middlebrook media. The proposed susceptibility breakpoint means that a larger proportion of clinical isolates is resistant to moxifloxacin prior to therapy. For patients infected with susceptible isolates, however, 800 mg a day should be examined for safety and efficacy for disseminated M. avium disease.


Antimicrobial Agents and Chemotherapy | 2013

Thioridazine Pharmacokinetic-Pharmacodynamic Parameters “Wobble” during Treatment of Tuberculosis: a Theoretical Basis for Shorter-Duration Curative Monotherapy with Congeners

Sandirai Musuka; Shashikant Srivastava; Chandima Wasana Siyambalapitiyage Dona; Claudia Meek; Richard Leff; Jotam G. Pasipanodya; Tawanda Gumbo

ABSTRACT Phenothiazines are being repurposed for treatment of tuberculosis. We examined time-kill curves of thioridazine and first-line drugs against log-growth-phase and semidormant bacilli under acidic conditions and nonreplicating persistent Mycobacterium tuberculosis. While both the potency and the efficacy of first-line drugs declined dramatically as M. tuberculosis replication rates decreased, those of thioridazine improved. The mutation prevalence to 3 times the thioridazine MIC was <1 × 10−11, better than for ≥2 first-line drugs combined. Hollow fiber system studies revealed that the relationship between sterilizing effect and pharmacodynamic indices (PDI) was characterized by an r2 of 0.88 for peak/MIC, an r2 of 0.47 for the area under the concentration-time curve (AUC) to MIC, and an r2 of 0.14 for the cumulative percentage of a 24-h period that the drug concentration exceeds the MIC under steady-state pharmacokinetic conditions (%TMIC) at the end of the first week. However, the PDI linked to effect “wobbled” as the duration of therapy increased, so that by the fourth week the r2 was 0.88 for AUC/MIC, 0.78 for %TMIC, and 0.72 for peak/MIC. This “wobble” has implications on general pharmacokinetic/pharmacodynamic theory, whereby efficacy is linked to only one of the three PDIs in deterministic models. The potency changed 8.9-fold from the first to the fourth weeks. The non-protein-bound AUC/MIC associated with maximal kill at the end of therapy was 50.53 (protein binding = 99.5%). This thioridazine exposure was calculated to extinguish all three M. tuberculosis metabolic populations in human lungs in only 42.9 days of monotherapy. However, this concentration exceeds the 2- to 8-mg/liter thioridazine concentration in serum known to be lethal to humans. Therefore, the way forward for phenothiazine monotherapy that also reduces therapy duration is via synthesis of less toxic congeners.


Current Pharmaceutical Design | 2011

In Vitro and In Vivo Modeling of Tuberculosis Drugs and its Impact on Optimization of Doses and Regimens

Shashikant Srivastava; Tawanda Gumbo

It has become increasingly clear that anti-tuberculosis regimens need optimization. Information gained using pharmacokinetics/ pharmacodynamics (PK/PD) methods in hollow fiber and animal model studies, in conjunction with Monte Carlo simulations, can be used to achieve this goal. PK/PD models of anti-tuberculosis drugs in hollow fibers, mice and guinea pigs have been remarkably concordant. Using exposures derived in these models it has been shown that the standard doses of pyrazinamide, rifampin, and ethambutol should be increased for a better efficacy, while doses of isoniazid need to be individualized. In addition, PK/PD driven doses have been proposed for new anti-tuberculosis agents such as moxifloxacin and PA-824.

Collaboration


Dive into the Shashikant Srivastava's collaboration.

Top Co-Authors

Avatar

Tawanda Gumbo

Baylor University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jotam G. Pasipanodya

Baylor University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Devyani Deshpande

Baylor University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Claudia Meek

Texas Tech University Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Richard Leff

Texas Tech University Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Carleton Sherman

Baylor University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Nicola M. Zetola

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Soumya Swaminathan

Indian Council of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Chawangwa Modongo

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Christopher Vinnard

Public Health Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge