Sunday, September 28, 2025

Antimicrobial Sensitivity Testing in Critical Care: Beyond the Numbers

 

Antimicrobial Sensitivity Testing in Critical Care: Beyond the Numbers - A Practical Guide for the Modern Intensivist

Dr Neeraj Manikath ,claude.ai

Abstract

Background: Antimicrobial sensitivity testing (AST) remains the cornerstone of targeted therapy in critically ill patients, yet interpretation extends far beyond simple susceptible/resistant classifications. The complex pathophysiology of critical illness, altered pharmacokinetics, and emergence of multidrug-resistant organisms demand sophisticated understanding of AST principles.

Objective: To provide critical care physicians with a comprehensive framework for interpreting AST results, incorporating pharmacokinetic-pharmacodynamic principles, clinical correlations, and practical pearls for optimizing antimicrobial therapy in the intensive care unit.

Methods: Comprehensive review of current literature, guidelines from major societies (CLSI, EUCAST, IDSA), and expert consensus on AST interpretation in critical care settings.

Results: This review addresses key concepts including minimum inhibitory concentration interpretation, breakpoint evolution, resistance mechanisms, and clinical correlation strategies. Practical pearls and common pitfalls are highlighted throughout.

Conclusions: Effective AST interpretation requires integration of microbiological data with patient-specific factors, understanding of resistance mechanisms, and appreciation of pharmacokinetic-pharmacodynamic principles to optimize outcomes in critically ill patients.

Keywords: antimicrobial sensitivity testing, critical care, minimum inhibitory concentration, breakpoints, pharmacokinetics, resistance mechanisms


Introduction

In the high-stakes environment of critical care medicine, antimicrobial therapy decisions can mean the difference between survival and mortality. While empirical therapy initiates treatment, antimicrobial sensitivity testing (AST) provides the roadmap for optimization. However, the interpretation of AST results in critically ill patients requires nuanced understanding that extends far beyond the binary susceptible/resistant paradigm.¹

The critically ill patient presents unique challenges: altered pharmacokinetics due to capillary leak, organ dysfunction, and extracorporeal therapies; increased infection severity requiring higher antimicrobial exposure; and higher prevalence of multidrug-resistant organisms.² These factors necessitate a sophisticated approach to AST interpretation that considers not just what the laboratory reports, but how to translate these findings into optimal clinical outcomes.

This review aims to equip the modern intensivist with practical tools for AST interpretation, incorporating recent advances in pharmacokinetic-pharmacodynamic modeling, resistance mechanism understanding, and clinical correlation strategies.


Fundamentals of Antimicrobial Sensitivity Testing

Minimum Inhibitory Concentration: The Foundation

The minimum inhibitory concentration (MIC) represents the lowest concentration of antimicrobial that inhibits visible bacterial growth after 18-24 hours of incubation.³ This seemingly simple concept forms the backbone of AST interpretation, yet its clinical application requires careful consideration.

Pearl #1: The MIC is a laboratory construct performed under standardized conditions that may not reflect the complex in vivo environment of critical illness. Temperature (35°C vs. physiologic 37°C), pH (7.2-7.4 vs. potentially acidotic tissue), oxygen tension, and protein binding all influence actual antimicrobial activity.⁴

Breakpoints: The Moving Targets

Clinical breakpoints categorize organisms as susceptible (S), intermediate (I), or resistant (R) based on achievable plasma concentrations with standard dosing.⁵ However, these breakpoints are not static and undergo regular revision based on:

  1. Pharmacokinetic-pharmacodynamic data
  2. Clinical outcome studies
  3. Resistance mechanism evolution
  4. Population pharmacokinetic modeling

Oyster #1: The "intermediate" category is often misunderstood. CLSI defines intermediate as "a category that includes isolates with antimicrobial agent MICs that approach usually attainable blood and tissue levels and for which response rates may be lower than for susceptible isolates."⁶ In critical care, this often translates to "may work with optimized dosing."

Evolution of Breakpoints: Clinical Implications

The evolution of breakpoints reflects our growing understanding of antimicrobial pharmacology. Notable examples include:

  • Fluoroquinolones against Enterobacteriaceae: Breakpoints were lowered due to recognition of treatment failures at previously "susceptible" MICs
  • Cephalosporins against Enterobacteriaceae: Introduction of ESBL screening changed interpretation paradigms
  • Vancomycin against Staphylococcus aureus: Elimination of the intermediate category reflected clinical outcome data⁷

Hack #1: Always check the year of your laboratory's breakpoint implementation. Older breakpoints may overestimate susceptibility for certain organism-antimicrobial combinations.


Resistance Mechanisms: The Clinical Detective Story

Understanding resistance mechanisms transforms AST interpretation from pattern recognition to mechanistic reasoning. This knowledge enables prediction of cross-resistance, selection of appropriate combination therapy, and anticipation of resistance development.

β-Lactam Resistance: The Great Deactivator

β-lactamases remain the most clinically significant resistance mechanism for gram-negative bacteria. Classification systems (Ambler, Bush-Jacoby) provide frameworks for understanding clinical implications.⁸

Extended-Spectrum β-Lactamases (ESBLs):

  • Clinical Pearl: ESBL-producing organisms should be reported as resistant to all penicillins, cephalosporins, and aztreonam, regardless of in vitro testing results
  • Mechanism: Hydrolysis of extended-spectrum cephalosporins and monobactams
  • Clinical Implication: Carbapenems remain first-line; ceftazidime-avibactam and ceftolozane-tazobactam show promise⁹

AmpC β-lactamases:

  • Clinical Pearl: Inducible AmpC can lead to treatment failure despite initial susceptibility
  • Organisms: Enterobacter spp., Citrobacter freundii, Serratia spp., Pseudomonas aeruginosa
  • Clinical Implication: Avoid extended-spectrum cephalosporins even if reported susceptible¹⁰

Carbapenemases:

  • KPC (Klebsiella pneumoniae carbapenemase): Inhibited by clavulanic acid
  • NDM (New Delhi metallo-β-lactamase): Requires zinc cofactor, inhibited by EDTA
  • OXA-48-like: Weak carbapenemase activity, may appear susceptible to carbapenems¹¹

Hack #2: Use the "carbapenem MIC creep" concept - rising carbapenem MICs (even within susceptible range) may herald emerging carbapenemase production before frank resistance appears.

Fluoroquinolone Resistance: The Multi-Target Problem

Fluoroquinolone resistance typically develops through:

  1. Target modification: DNA gyrase (gyrA, gyrB) and topoisomerase IV (parC, parE) mutations
  2. Efflux pumps: Particularly in Pseudomonas and Acinetobacter
  3. Plasmid-mediated resistance: qnr genes, AAC(6')-Ib-cr¹²

Clinical Pearl #2: Cross-resistance between fluoroquinolones is common but not absolute. Ciprofloxacin resistance doesn't always predict levofloxacin resistance, particularly in Streptococcus pneumoniae.

Aminoglycoside Resistance: The Modifier Enzymes

Aminoglycoside-modifying enzymes (AMEs) confer resistance through acetylation, phosphorylation, or adenylation. Clinical implications include:

  • AAC(6')-Ie: Confers resistance to amikacin, netilmicin, and tobramycin but not gentamicin
  • APH(3')-VIa: Confers high-level gentamicin resistance in enterococci
  • 16S rRNA methylases: Confer pan-aminoglycoside resistance¹³

Oyster #2: Aminoglycoside susceptibility in gram-positive cocci requires both screen-positive (low-level resistance overcome by synergy) and high-level resistance testing. This distinction is crucial for endocarditis therapy.


Pharmacokinetic-Pharmacodynamic Principles in AST Interpretation

The integration of PK-PD principles transforms static MIC values into dynamic predictors of clinical outcome. Understanding these relationships is essential for optimizing therapy in critically ill patients.

Time-Dependent Killing: β-lactams and Glycopeptides

For time-dependent antimicrobials, efficacy correlates with the percentage of dosing interval that free drug concentrations exceed the MIC (fT>MIC).

Clinical Targets:

  • Penicillins: 40-50% fT>MIC for bacteriostatic effect, 60-70% for bactericidal effect
  • Cephalosporins: 60-70% fT>MIC
  • Carbapenems: 30-40% fT>MIC (due to post-antibiotic effect)
  • Vancomycin: AUC₀₋₂₄/MIC ratio 400-600¹⁴

Hack #3: For β-lactams against organisms with MICs at the susceptible breakpoint, consider extended or continuous infusion to maximize fT>MIC, particularly in patients with augmented renal clearance.

Concentration-Dependent Killing: Aminoglycosides and Fluoroquinolones

Efficacy correlates with peak concentration relative to MIC (Cₘₐₓ/MIC) or area under the curve relative to MIC (AUC₀₋₂₄/MIC).

Clinical Targets:

  • Aminoglycosides: Cₘₐₓ/MIC ratio 8-10 for gram-negative bacteria, 10-12 for gram-positive
  • Fluoroquinolones: AUC₀₋₂₄/MIC ratio 100-125 for gram-negative bacteria¹⁵

Pearl #3: In patients with altered volume of distribution (capillary leak, fluid resuscitation), aminoglycoside dosing based on actual body weight may be insufficient to achieve target Cₘₐₓ/MIC ratios.


Special Considerations in Critical Care

Altered Pharmacokinetics in Critical Illness

Critical illness profoundly affects antimicrobial pharmacokinetics through multiple mechanisms:

Increased Volume of Distribution:

  • Capillary leak syndrome
  • Fluid resuscitation
  • Hypoalbuminemia
  • Clinical Impact: Reduced peak concentrations for concentration-dependent antimicrobials¹⁶

Altered Clearance:

  • Augmented renal clearance (ARC) in early sepsis
  • Acute kidney injury in later stages
  • Hepatic dysfunction
  • Clinical Impact: Subtherapeutic levels despite standard dosing¹⁷

Protein Binding Changes:

  • Hypoalbuminemia increases free fraction
  • Acute-phase proteins may increase binding
  • Clinical Impact: Complex effects on antimicrobial activity¹⁸

Hack #4: Consider therapeutic drug monitoring for narrow therapeutic index antimicrobials (vancomycin, aminoglycosides) and those with wide PK variability in critical illness (β-lactams, fluoroquinolones).

Tissue Penetration Considerations

AST is performed in broth media, but infections occur in tissues with varying penetration characteristics:

CNS Infections:

  • Only free, non-protein-bound antimicrobial crosses blood-brain barrier
  • Inflammation increases penetration but may not normalize ratios
  • Clinical Pearl: Use higher susceptible breakpoints when available for CNS infections¹⁹

Pulmonary Infections:

  • Epithelial lining fluid concentrations vary widely between antimicrobials
  • Aminoglycosides have poor lung penetration
  • Fluoroquinolones and lincosamides achieve excellent lung levels²⁰

Intra-abdominal Infections:

  • Anaerobic environment may reduce antimicrobial activity
  • Abscess penetration is limited for many antimicrobials
  • Clinical Pearl: Consider source control as primary intervention when AST shows borderline susceptibility²¹

Biofilm-Associated Infections

Device-associated infections often involve biofilm formation, which significantly alters antimicrobial susceptibility:

  • Reduced penetration through extracellular matrix
  • Altered physiology of biofilm-embedded bacteria
  • Persister cells tolerant to antimicrobial exposure
  • Clinical Implication: MICs may underestimate treatment difficulty²²

Oyster #3: Standard AST doesn't detect biofilm-associated resistance. Consider device removal even when organism appears "susceptible" if clinical response is poor.


Practical Pearls and Clinical Correlations

The Art of AST Interpretation

Pearl #4: Always correlate AST results with clinical presentation. A "susceptible" organism causing treatment failure suggests:

  • Inadequate source control
  • Poor tissue penetration
  • Suboptimal dosing
  • Alternative diagnosis
  • Laboratory error

Pearl #5: Resist the temptation to "follow the antibiogram." Local resistance patterns inform empirical therapy but individual patient AST should guide definitive treatment.

Common Interpretation Pitfalls

Pitfall #1: The Heteroresistance Trap Some organisms contain subpopulations with different susceptibilities. Standard AST may miss minority resistant populations that emerge under selective pressure.

  • Most Common: Vancomycin-intermediate S. aureus (VISA)
  • Clinical Clue: Rising vancomycin MICs over time²³

Pitfall #2: The Disk Diffusion Deception Zone diameters can be affected by:

  • Disk storage conditions
  • Inoculum density
  • Medium depth
  • Clinical Impact: May miss borderline resistance²⁴

Pitfall #3: The Combination Therapy Confusion AST typically tests single antimicrobials, but synergy testing is limited. Clinical outcomes with combination therapy may exceed predictions from individual susceptibilities.

Modern Molecular Methods

Rapid molecular diagnostics are revolutionizing AST interpretation:

Advantages:

  • Rapid turnaround time (1-6 hours vs. 24-48 hours)
  • Direct detection of resistance genes
  • Species identification without culture

Limitations:

  • Limited resistance gene panels
  • Cannot detect novel resistance mechanisms
  • No MIC values for dosing optimization²⁵

Hack #5: Use molecular results for rapid de-escalation and targeted empirical therapy, but confirm with conventional AST for optimization.


Resistance Surveillance and Stewardship

Local Antibiograms: The Institutional Compass

Understanding your local resistance patterns is crucial for:

  • Empirical therapy selection
  • Recognizing unusual resistance patterns
  • Tracking resistance trends
  • Informing infection control measures²⁶

Pearl #6: Pay attention to resistance trend changes over time. A sudden increase in carbapenem resistance may herald a new resistance mechanism before individual cases are recognized.

Antimicrobial Stewardship Integration

AST interpretation should be integrated into stewardship programs:

De-escalation Strategies:

  • Narrow spectrum based on final AST
  • Switch from IV to oral when appropriate
  • Optimize dosing based on PK-PD principles

Duration Optimization:

  • Shorter courses for uncomplicated infections
  • Biomarker guidance (procalcitonin) when appropriate
  • Clinical response assessment²⁷

Emerging Technologies and Future Directions

Rapid Phenotypic Methods

New technologies promise to accelerate AST results:

  • Flow cytometry-based systems: Results in 4-6 hours
  • Digital microscopy: Real-time growth monitoring
  • Metabolic activity assays: Detect growth inhibition rapidly²⁸

Artificial Intelligence Applications

Machine learning approaches may enhance AST interpretation:

  • Pattern recognition for resistance mechanisms
  • Prediction of clinical outcomes
  • Optimization of dosing regimens
  • Integration of multi-omics data²⁹

Precision Medicine Approaches

Future AST interpretation may incorporate:

  • Individual patient pharmacokinetics
  • Host immune status
  • Pathogen virulence factors
  • Site-of-infection characteristics³⁰

Case-Based Applications

Case 1: The Vancomycin Conundrum

Clinical Scenario: 65-year-old post-cardiac surgery patient with MRSA bacteremia. Blood cultures positive for S. aureus with vancomycin MIC = 2 mg/L (susceptible).

AST Interpretation Considerations:

  • MIC at upper limit of susceptible range
  • Risk of heteroresistance (hVISA)
  • Pharmacokinetic challenges in cardiac surgery patients
  • Alternative agents (daptomycin, linezolid, ceftaroline) may be preferred³¹

Clinical Pearl #7: For MRSA with vancomycin MIC ≥1.5 mg/L, consider alternative agents regardless of "susceptible" designation.

Case 2: The Pseudomonas Predicament

Clinical Scenario: 45-year-old burn patient with P. aeruginosa pneumonia. AST shows: piperacillin-tazobactam (susceptible), cefepime (intermediate), meropenem (susceptible), ciprofloxacin (resistant), tobramycin (susceptible).

AST Interpretation Considerations:

  • Risk of inducible AmpC with extended-spectrum β-lactams
  • Meropenem preferred despite piperacillin-tazobactam susceptibility
  • Combination therapy consideration for severe infection
  • Burn patients often have altered pharmacokinetics³²

Hack #6: For serious P. aeruginosa infections, combination therapy with two mechanistically different antimicrobials may improve outcomes even when monotherapy appears adequate based on AST.


Quality Assurance and Laboratory Communication

Ensuring AST Accuracy

Critical care physicians should understand laboratory quality measures:

Quality Control Strains:

  • ATCC reference strains tested daily
  • Expected MIC ranges for each antimicrobial
  • Corrective actions for out-of-range results³³

Proficiency Testing:

  • External quality assessment programs
  • Inter-laboratory comparison
  • Trending of performance metrics

Effective Laboratory Communication

When to Call the Lab:

  • Unusual resistance patterns
  • Discrepancy between clinical response and AST
  • Questions about methodology
  • Requests for additional testing

Hack #7: Develop a relationship with your clinical microbiology team. Their expertise in resistance mechanisms and local epidemiology is invaluable for complex cases.


Conclusions and Clinical Recommendations

Antimicrobial sensitivity testing interpretation in critical care requires integration of microbiological principles, pharmacokinetic-pharmacodynamic understanding, and clinical correlation. Key recommendations include:

  1. Look beyond S/I/R categories - Consider MIC values, resistance mechanisms, and PK-PD principles
  2. Understand your patient - Critical illness alters pharmacokinetics and may require dosing adjustments
  3. Know your local epidemiology - Resistance patterns inform both empirical and targeted therapy
  4. Integrate with stewardship - Use AST for optimization, not just selection
  5. Communicate with your laboratory - Leverage microbiologist expertise for complex cases
  6. Stay current - Breakpoints, resistance mechanisms, and technologies continue to evolve

The future of AST interpretation lies in personalized medicine approaches that consider individual patient factors, pathogen characteristics, and site-of-infection physiology. Until these tools become widely available, thoughtful application of current principles will continue to optimize outcomes for critically ill patients.


References

  1. Rhodes A, Evans LE, Alhazzani W, et al. Surviving sepsis campaign: international guidelines for management of sepsis and septic shock: 2016. Intensive Care Med. 2017;43(3):304-377.

  2. Roberts JA, Paul SK, Akova M, et al. DALI: defining antibiotic levels in intensive care unit patients: are current β-lactam antibiotic doses sufficient for critically ill patients? Clin Infect Dis. 2014;58(8):1072-1083.

  3. Clinical and Laboratory Standards Institute. Performance Standards for Antimicrobial Susceptibility Testing. 33rd ed. CLSI supplement M100. Wayne, PA: CLSI; 2023.

  4. Mouton JW, Muller AE, Canton R, et al. MIC-based dose adjustment: facts and fables. J Antimicrob Chemother. 2018;73(3):564-568.

  5. European Committee on Antimicrobial Susceptibility Testing. Breakpoint tables for interpretation of MICs and zone diameters. Version 13.0. 2023.

  6. Kahlmeter G, Brown DF, Goldstein FW, et al. European harmonization of MIC breakpoints for antimicrobial susceptibility testing of bacteria. J Antimicrob Chemother. 2003;52(2):145-148.

  7. Rybak MJ, Le J, Lodise TP, et al. Therapeutic monitoring of vancomycin for serious methicillin-resistant Staphylococcus aureus infections: A revised consensus guideline and review by the American Society of Health-System Pharmacists. Am J Health Syst Pharm. 2020;77(11):835-864.

  8. Bush K, Bradford PA. Epidemiology of β-lactamase-producing pathogens. Clin Microbiol Rev. 2020;33(2):e00047-19.

  9. Karaiskos I, Lagou S, Pontikis K, et al. Ceftazidime/avibactam and ceftolozane/tazobactam for the treatment of multidrug-resistant gram-negative bacterial infections. Expert Rev Anti Infect Ther. 2019;17(12):983-995.

  10. Harris PN, Tambyah PA, Lye DC, et al. Effect of piperacillin-tazobactam vs meropenem on 30-day mortality for patients with E coli or Klebsiella pneumoniae bloodstream infection and ceftriaxone resistance. JAMA. 2018;320(10):984-994.

  11. Nordmann P, Naas T, Poirel L. Global spread of carbapenemase-producing Enterobacteriaceae. Emerg Infect Dis. 2011;17(10):1791-1798.

  12. Redgrave LS, Sutton SB, Webber MA, et al. Fluoroquinolone resistance: mechanisms, impact on bacteria, and role in evolutionary success. Trends Microbiol. 2014;22(8):438-445.

  13. Ramirez MS, Tolmasky ME. Aminoglycoside modifying enzymes. Drug Resist Updat. 2010;13(6):151-171.

  14. Abdul-Aziz MH, Sulaiman H, Mat-Nor MB, et al. Beta-lactam infusion in severe sepsis (BLISS): a prospective, two-centre, open-labelled randomised controlled trial. Intensive Care Med. 2016;42(10):1535-1545.

  15. Drusano GL. Antimicrobial pharmacodynamics: critical interactions of 'bug and drug'. Nat Rev Microbiol. 2004;2(4):289-300.

  16. Heffernan AJ, Sime FB, Lipman J, et al. Individualising therapy to minimize bacterial multidrug resistance. Drugs. 2018;78(3):267-292.

  17. Udy AA, Varghese JM, Altukroni M, et al. Subtherapeutic initial β-lactam concentrations in select critically ill patients: association between augmented renal clearance and low trough drug concentrations. Chest. 2012;142(1):30-39.

  18. Wong G, Briscoe S, McWhinney B, et al. Protein binding of β-lactam antibiotics in critically ill patients: can we successfully predict unbound concentrations? Antimicrob Agents Chemother. 2013;57(12):6165-6170.

  19. Nau R, Sörgel F, Eiffert H. Penetration of drugs through the blood-cerebrospinal fluid/blood-brain barrier for treatment of central nervous system infections. Clin Microbiol Rev. 2010;23(4):858-883.

  20. Rodvold KA, George JM, Yoo L. Penetration of anti-infective agents into pulmonary epithelial lining fluid: focus on antibacterial agents. Clin Pharmacokinet. 2011;50(10):637-664.

  21. Solomkin JS, Mazuski JE, Bradley JS, et al. Diagnosis and management of complicated intra-abdominal infection in adults and children: guidelines by the Surgical Infection Society and the Infectious Diseases Society of America. Clin Infect Dis. 2010;50(2):133-164.

  22. Høiby N, Bjarnsholt T, Moser C, et al. ESCMID guideline for the diagnosis and treatment of biofilm infections 2014. Clin Microbiol Infect. 2015;21(Suppl 1):S1-S25.

  23. Howden BP, Davies JK, Johnson PD, et al. Reduced vancomycin susceptibility in Staphylococcus aureus, including vancomycin-intermediate and heterogeneous vancomycin-intermediate strains: resistance mechanisms, laboratory detection, and clinical implications. Clin Microbiol Rev. 2010;23(1):99-139.

  24. Jorgensen JH, Ferraro MJ. Antimicrobial susceptibility testing: a review of general principles and contemporary practices. Clin Infect Dis. 2009;49(11):1749-1755.

  25. Opota O, Croxatto A, Prod'hom G, et al. Blood culture-based diagnosis of bacteraemia: state of the art. Clin Microbiol Infect. 2015;21(4):313-322.

  26. Hindler JF, Stelling J. Analysis and presentation of cumulative antibiograms: a new consensus guideline from the Clinical and Laboratory Standards Institute. Clin Infect Dis. 2007;44(6):867-873.

  27. Barlam TF, Cosgrove SE, Abbo LM, et al. Implementing an antibiotic stewardship program: guidelines by the Infectious Diseases Society of America and the Society for Healthcare Epidemiology of America. Clin Infect Dis. 2016;62(10):e51-e77.

  28. Idelevich EA, Becker K. How to accelerate antimicrobial susceptibility testing. Clin Microbiol Infect. 2019;25(11):1347-1355.

  29. Weis CV, Jutzeler CR, Borgwardt K. Machine learning for microbial identification and antimicrobial susceptibility testing on MALDI-TOF mass spectra: a systematic review. Clin Microbiol Infect. 2020;26(10):1310-1317.

  30. Doern CD. The slow march toward rapid phenotypic antimicrobial susceptibility testing: are we there yet? J Clin Microbiol. 2018;56(4):e01999-17.

  31. Liu C, Bayer A, Cosgrove SE, et al. Clinical practice guidelines by the Infectious Diseases Society of America for the treatment of methicillin-resistant Staphylococcus aureus infections in adults and children. Clin Infect Dis. 2011;52(3):e18-e55.

  32. Blanchet B, Jullien V, Vinsonneau C, et al. Influence of burns on pharmacokinetics and pharmacodynamics of drugs used in the care of burn patients. Clin Pharmacokinet. 2008;47(10):635-654.

  33. Clinical and Laboratory Standards Institute. Quality Control for Commercial Microbiology Transport Systems. 2nd ed. CLSI standard M40. Wayne, PA: CLSI; 2014.

Conflict of Interest: None declared

Funding: None

Ethics: Not applicable (review article)

No comments:

Post a Comment

Precision Fluid Therapy in Shock: Integrating Dynamic Assessment, Organ Congestion Monitoring

  Precision Fluid Therapy in Shock: Integrating Dynamic Assessment, Organ Congestion Monitoring, and Artificial Intelligence Dr Neeraj Manik...