0% found this document useful (0 votes)
4 views

M Tech in Pharmaceutical Biotechnology Syllabus

The document outlines the syllabus structure for the M.Tech in Pharmaceutical Biotechnology program, detailing courses, subjects, hours per week, marks, and credits across four semesters. It includes core courses such as Pharmaceutical Biotechnology, Advances in Recombinant DNA Technology, and Research Methodology, as well as a list of electives. Additionally, it provides a comprehensive overview of core subjects and elective topics, emphasizing practical applications and research methodologies in the field.

Uploaded by

kundaladevi922
Copyright
© © All Rights Reserved
Available Formats
Download as PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
4 views

M Tech in Pharmaceutical Biotechnology Syllabus

The document outlines the syllabus structure for the M.Tech in Pharmaceutical Biotechnology program, detailing courses, subjects, hours per week, marks, and credits across four semesters. It includes core courses such as Pharmaceutical Biotechnology, Advances in Recombinant DNA Technology, and Research Methodology, as well as a list of electives. Additionally, it provides a comprehensive overview of core subjects and elective topics, emphasizing practical applications and research methodologies in the field.

Uploaded by

kundaladevi922
Copyright
© © All Rights Reserved
Available Formats
Download as PDF, TXT or read online on Scribd
You are on page 1/ 13

Syllabus Structure- M Tech.

Pharmaceutical
Biotechnology

Semester- I

No. Contents Course Subjects Hours/Week Marks Credits


Code (L+T)
PBT 2101 Pharmaceutical
1. Core I Biotechnology (2+1) 50 3
I
PBT 2102 Advances in
2. Core II Recombinant (2+1) 50 3
DNA
Technology
PBT 2106 Make-up
subject-1
Fundamentals
of
3. Core III Microbiology, (2+1) 50 3
Molecular
Biology and
Chemical
Engineering
4. Elective I (2+1) 50 3

5. Elective II (2+1) 50 3

Seminar PBT 2114 Project – I


6. and (Seminar and 6 50 3
Critical Critical
Review
Review)
PHP 2505 Laboratory I-
7 Practical Instrumental 6 50 3
Methods of
Analysis
8 Research I PBT 2115 Research I 12 100 6

TOTAL 39 450 3
Semester- II

No. Contents Course Subjects Hours/Week Marks Credits


Code (L+T)
PBT 2103 Research
1. Core I (2+1) 50 3
Methodology
PBT 2104 Pharmaceutical
2. Core II Biotechnology (2+1) 50 3
II
PBT 2105 Advanced
3. Core III Analytical (2+1) 50 3
Tools in
Biotechnology
4. Elective III (2+1) 50 3

5. Elective IV (2+1) 50 3
PBL 2002 Laboratory –II
6. Practical (Microbiology 6 50 3
and Molecular
biology)
PBT 2116 Research II
7. Research II 18 150 9

TOTAL 39 450 27

Semester- III

No. Course Hours/Week Marks Credits

1. In plant 40 (15 450 30


training Weeks)

TOTAL 40 450 30

Semester- IV

No. Course Hours/Week Marks Credits


Research,
1. Thesis and 40 450 30
Open
defense

TOTAL 40 450 30

Sem III and Sem IV Evaluation will conducted be at end of IV semester.


List of Electives:
1. PBT 2121 Advanced Biochemistry
2. PBT 2122 Advanced Bioinformatics
3. PBT 2123 Biostatistics
4. PHT 2004 Drug Metabolism
5. PBT 2124 Environmental Biotechnology
6. PBT 2125 Immunology
7. PBT 2129 Vaccines and Immunotherapy
8. PHT 2002 Intellectual Property Rights and Patent Filing
9. PHT 2005 Molecular Biology
10. PBT 2126 Protein and Nucleic Acid Formulation Development
11. PHT 2302 Pharmacology, Toxicology and Therapeutics
12. PBT 2127 Process Biotechnology
13. PBT 2128 Tissue Engineering and Biopolymers
14. PHT 2107 Targeted Drug Delivery Systems
15. BST 2102 Unit Operation in Bioprocesses
16. CEE 2003 Environmental Biotechnology
17. BSE 2110 Biocatalysis and Green Technology
18. FDT 2005 Carbohydrate Chemistry & Technology
19. FDT 2072 Nutritional Genomics

Subject wise Syllabus


I. Core subjects
1. Pharmaceutical Biotechnology I
1. Biotechnology in the Pharmaceutical Industry (Pre-biotechnology products, impact of
biotechnology, post-biotechnology products: biologics and biopharmaceuticals)
2. Genetic manipulation methods
3. Fermentation technology
4. Scale-up process (Inoculum: preparation and development of inoculum for industrial
fermentation, optimization of the fermentation process (pH, temperature, and oxygen
requirements, Determination of the optimized feeding regimen and biomass
quantification
5. Improvement of selected microorganism with increased productivity of the fermented
products
6. Fermentation process: Batch and continuous fermentation and fermenters, Fermentation
products in Pharmaceutical industry: Antibodies, Therapeutic proteins, Vitamins, Amino
acids, Monoclonal Antibodies)

2. Advances in Recombinant DNA Technology


1. Vectors: Cloning vectors: Plasmids, Lambda phages, single stranded DNA vectors (M13,
fd, f1); Cosmids, Phasmids and Phagemids, YACs, BACs, PACs; Plant Transformation
vectors: Introduction to Ti, Ri plasmids and BIBACs; Expression Vectors for high level
protein expression
2. Cloning strategies: Vector preparation and diverse cloning strategies viz. blunt end
cloning, directional cloning, TA-cloning of PCR products, linkers and adaptors based
cloning methodologies
3. E. coli transformation: Chemical transformation and Electroporation
4. Selection and screening of recombinant transformants: Introduction to marker and
reporter genes and selection strategies
5. Labeling and detection of nucleic acid sequences: End-Labeling (3’- and 5’-), Random
priming and Nick translation using radioactive non-radioactive labeling techniques
6. Genomic DNA libraries: Procedures for Partial, Representative, Enriched, Large-insert
DNA libraries, Half-arm cloning, cDNA libraries: Prominent Adapters/Linkers based
directional cloning
7. Gene therapy for genetic diseases

3. Research Methodology
Research
1. Meaning of Research, Purpose of Research, Types of Research (Educational, Clinical,
Experimental, Historical, Descriptive, Basic applied and Patent Oriented Research) –
Objective of research-
2. Literature survey – Use of Library, Books, & Journals – Medline – Internet, getting
patents and reprints of articles as sources for literature survey.
3. Selecting a problem and preparing research proposal for different types of research
mentioned above.
4. Methods and tools used in Research
• Qualitative studies, Quantitative Studies
• Simple data organization, Descriptive data analysis
• Limitations and sources of Error
• Inquiries in form of Questionnaire, Opinionnaire or by interview
• Statistical analysis of data including variance, standard deviation, students ‘t’ test and
annova, correlation data and its interpretation, computer data analysis,
5. Documentation
• “How” of Documentation
• Techniques of Documentation
• Importance of Documentation
• Uses of computer packages in Documentation
6. The Research Report / Paper writing / thesis writing
• Different parts of the Research paper
• Title – Title of project with author’s name
• Abstract – Statement of the problem Background list in brief and purpose and scope
• Key-words-
• Methodology-Subject, Apparatus / Instrumentation, (if necessary) and procedure
• Results – tables, Graphs, Figures, and statistical presentation
• Discussion – Support or non- support of hypothesis – practical & theoretical implications,
conclusions
• Acknowledgements
• References
• Errata
• Importance of spell check for Entire project
• Use of footnotes
7. Presentation (Specially for oral)
• Importance, types, different skills
• Content of presentation, format of model, Introduction and ending
• Posture, Genstures, Eye contact, facial expressions stage fright
• Volume- pitch, speed, pauses & language
• Visual aids and seating
• Questionnaire
8. Protection of patents and trade marks, Designs and copyrights
• The patent system in India – Present status Intellectual property Rights (IPR), Future
changes expected in Indian Patents
• Advantages
• The Science in Law, Turimetrics (Introduction)
• What may be patented
• Who may apply for patent
• Preparation of patent proposal
• Registration of patent in foreign countries and vice-versa
9. Sources for procurement of Research Grants
10. Industrial- Institution Interaction
• Industrial projects – Their feasibility reports
• Research in Education – Johan V. Best James V. Kahn
• Presentation skills- Michael Halton- Indian Society for Institute Education
• A Practical Introduction to copy right – Gavin Mcfarlane
• Thesis projects in Science and Engineering – Richard M. Davis
• Scientists in legal system – Ann labor science
• Thesis and Assignment writing – Jonathan Anderson
• Writing a technical paper- Donald Menzel
• Effective Business Report writing – Leland Brown
• Protection of Industrial property rights- Purushottam Das and Gokul Das
• Spelling for the million – Edna furmess
• Preparing for publication – King Edwards Hospital fund for London
• Information technology – The Hindu speaks
• Documentation – Genesis & Development 3792
• Manual for evaluation of Industrial projects – United Nations
• Manual for the preparation of Industrial feasibility studies

4. Pharmaceutical Biotechnology II
1. Animal Cell Culture: Historical Background, Importance of and progress in Animal Cell
Culture, Technology, Biology of Animal Cell; Cellular Interactions, Importance of Serum
and Serum Free Media, Culturing and Sub-Culturing of Animal Cells, InVitro
Transformation of Animal Cells, Cell Differentiation & Cell Movement, Cloning of
Animal Cells, Cell Line Preservation, Cell Line Characterization, Chromosome
Spreading and Karyotype Analysis, Mycoplasma: Detection and Control, Monoclonal
Antibody Production, Insect Cell Culture: An Overview
2. Plant cell culture: History and evolution, Basics of aseptic culture, In vitro propagation,
use of plant growth regulators in tissue culture, plant regeneration, organogenesis,
somatic embryogenesis, protoplast isolation and culture, somaclonal variation, in vitro
mutagenesis, in vitro selection, secondary metabolite production and cell transformation
techniques (including protoplast fusion, direct DNA uptake and plant/ bacterial co-
cultivation), use of in vitro techniques for crop improvement.
3. Omics: Proteomics, Genomics and Metabolomics: Introduction to the definitions of
various ‘omics’, introduction to the general field of genomics and proteomics,
introduction to some methods used in analyzing gene expression at the mRNA and
protein level, basic principles of DNA/Protein microarrays and their applications.
4. Physical aspects of the genome. Construction and study of various types of genome maps
and large-scale sequencing. The human genome project and the plant genome program.
Structural genomics and gene discovery, isolation, localization and characterization.
Developing diagnostic tests for plant, animal and human diseases. Identification of
biomarkers. Finding genetic markers for plant breeding purposes. Environmental impacts
on gene expression. Protein complex structures and amino acids. Protein shapes as
affecting its function. Amino acid sequencing. Cellular proteome changes in response to
environmental and neighbouring cells conditions. Cataloguing the proteins produced by
different cells. Discovering the function of a protein. Determining three-dimensional
structure of proteins. Protein crystallography.
5. Integrons and transposons
6. Regulatory aspects of biotechnology based products

5. Advanced Analytical Tools in Biotechnology


1. Diagnostic Methods - Molecular Methods: Isolation and purification of nucleic acid and
protein, Electophoresis and visualisation of nucleic acid and protein, Blotting techniques,
Sequencing and amplification techniques, PCR and related techniques
2. Genomic and Post-Genomic Analytical Biotechnology: Gene purification and sequencing,
Protein sequencing and purification, The goal and applications of genomics and proteomics,
Techniques in use for gene and protein analysis, e.g. crystallography, magnetic resonance
3. Immunological Methods: Antibody production and labeling, Immunochemical techniques
for in situ analyses (ICC and IHC), Immunochemical techniques for measurement (ELISA,
etc), Immunochemical techniques for separation (Immunoprecipitation, etc)
4. Introduction to Bioinformatics: organization of biological data, databases (raw and
processed), quering in databases, primer designing, gene finding, motif finding, sequence
alignment, protein sequence analysis

6. Fundamentals of Microbiology, Molecular Biology and Chemical Engineering


1. Fundamentals of Microbiology (10 lectures): Microbes – types, size shape and arrangement
of bacterial cells, Nutritional requirements – Common ingredients, culture media and types
of media, Sterilization – Importance and various methods of sterilization, Cultivation and
Preservation of microorganisms – Isolation, pure culture, study of cultural characteristics
and methods of preservation, Measurement of microbial growth – Total count and viable
count methods, Preparation of microbes for microscopic observation – Compound
microscope, stains used, simple staining, differential staining and special staining
techniques.
2. Fundamentals of Molecular Biology (10 lectures): The beginnings of Molecular biology,
The structure of DNA, Genome organization: Prokaryotes and Eukaryotes., The Versatility
of RNA: Types of RNA and their role, DNA replication: Prokaryotic and Eukaryotic,
Overview of Transcription in prokaryotes and eukaryotes, From Gene to Protein: Genetic
code and Translation, Recombinant DNA technology: An introduction, molecular cloning
and some tools for analyzing gene expression
3. Fundamentals of Chemical Engineering (10 lectures): Transport phenomenon, Heat transfer,
Mass transfer, Process and equipment design for various operations in processing of
pharmaceutical biotechnology based products and discussions on scale-up of operations;
Prediction of freezing, heating and drying times
III. Electives
1. Immunology
1. Immunology as a science, module overview, practical application of the module
2. Immunity: basic definitions, types of immunity, organs involved, cells of the immune
system, humoral v/s cellular immunity
3. Innate immunity: meaning, cells ‘producing’ innate immunity, non-cellular innate immunity
4. Adaptive immunity: meaning, cells bringing about specific immunity, production of a
specific response
5. Adaptive immunity 2: signaling, steps in the development of a nonself destructive cellular
specific immunity, steps in the development of a nonself destructive humoral specific
immunity
6. Adaptive immunity 3: cells and antibodies adcc, failure of specific immunity development
7. Antibodies & its diversity: types, development of specific antibodies in the body: vdj
recombination
8. Fine balance in immunity: th1 v/s th2 response
9. Mucosal immunology: mediation of immunology on mucosal surfaces, importance
10. Immunological response to disease (example study): components of an immunological
response, th1 response type disease, th2 response type disease, alternatively activated
macrophages, disease with primary response
11. Immunological pathogenesis: hyper sensitivity, auto immunity, allergy
12. Disease of the immune system: Overview
13. Cells of the immune system: identification, laboratory culture, primary culture, cell lines
14. Immunological techniques: diagnostic tests, basic/classical/common, elisa/ria call types,
western blot
15. Immune suppression
16. Graft Vs host diseases
17. HLA, HLA typing and applications including organ transplantation
18. Gut microbiota and Gut immunology

2. Vaccines and Immunotherapy


Syllabus to be framed

3. Protein Nucleic And Acid Formulation Development


1. Protein engineering
2. Nucleic acids and proteins: physicochemical properties and stabilization
3. Formulation aspects
4. Mechanisms of action
5. Characterization: Raman, Mass Spectrometry, Atomic Force and Scanning Electron
Microscopy (AFM and SEM), Confocal microscopy, Flow cytometry, Capillary DSC,
MALDI-TOF, circular dichroism
6. Biophysical techniques: pharmacokinetics/pharmacodynamics
7. In vitro studies: cellular trafficking
8. Biosimilars

4. Advanced Biochemistry
1. Proteins: Structures – primary, secondary, tertiary, motifs, structural and functional
domains, protein families and macromolecular assemblies.
2. Mechanisms for regulating protein function: Protein-protein interactions, interaction with
ligands, Ca¬+2 and GTP as modulators, cyclic phosphorylation and dephosphorylation,
proteolytic cleavage.
3. Purification and characterization of proteins: Electrophoresis, ultracentrifugation and
liquid chromatography, use of biological assays, use of radioisotopes and MS, X-ray
crystallography, NMR and Homology modeling, amino acid analysis, cleavage of
peptides, protein sequencing.
4. Protein biosynthesis: Translation machinery in prokaryotic and eukaryotic systems,
comparison of similarities and differences.
5. DNA and nucleic acids: DNA, RNA structure, nomenclature, double helix,
conformations, higher order packing and architecture of DNA, transcription and
replication of DNA – mechanisms in prokaryotic and eukaryotic systems, DNA repair
mechanisms.
6. Carbohydrates: Mono, di and polysaccharides and their nomenclature, stereochemistry,
linkages, conjugates of carbohydrates with other molecules - glycoproteins, glycolipids,
proteoglycans, lipopolysaccharides and their biological roles.
7. Lipids: Classification, nomenclature, stereochemistry, storage lipids, membrane lipids,
lipids as second messengers and cofactors, biological role of lipids

5.Advanced Bioinformatics
1. Motif and cis-Regulatory Module (CRM) Modeling: learning motif models, learning
models of cis-regulatory modules, Gibbs sampling, Dirichlet priors, parameter tying,
heuristic search, HMM structure search, sequence entropy and mutual information,
duration modeling, semi-Markov models
2. Gene Finding: the gene finding task, maximal dependence decomposition, interpolated
Markov models, back-off models, pairwise HMMs, Genscan, Twinscan, SLAM
3. RNA-Seq: RNA-Seq technology, transcript quantification with RNA-Seq
4. RNA Analysis: predicting RNA secondary structure, Nussinov/energy-minimization
algorithms, stochastic context free grammars, Inside/Inside-Outside/CYK algorithms,
searching sequences for a given RNA secondary structure, RSEARCH, RNA gene
recognition via comparative sequence analysis, microRNA gene/target prediction
5. Large-Scale and Whole-Genome Sequence Alignment: large-scale alignment, whole-
genome alignment, parametric alignment, suffix trees, locality sensitive hashing, k-mer
tries, sparse dynamic programming, longest increasing subsequence problem, Markov
random fields, MUMmer, LAGAN/MLAGAN, Mauve, Mercator
6. Biological network inference and evolution: Network inference, models of biological
network evolution, network alignment
7. Genotype Analysis: haplotype inference, genome-wide association studies (GWAS),
quantitative trait loci (QTL) mapping
8. Protein Structure Prediction: secondary structure prediction, threading, branch and bound
search, ROSETTA

6. Bio-statistics
1. Application of Statistics, bioinformatics and experimental design to biotech processes:
Sampling procedures, populations; types of data, data organization and presentation.
2. Correlation and Regression, linear and quadratic regression Analysis of variance.
3. Correlation coefficient; regression analysis; multivariate analysis; principal component
analysis. Probability. Probability distribution.
4. Testing of hypothesis. Experimental design and factorial design.
5. Concepts and use of software. RSM and ANN techniques for optimization of
fermentative processes

7. Drug Metabolism
1. Introduction to the different pathways of drug metabolism: Phase I and II reactions, sites
of drug metabolism, subcellular localization of drug metabolizing enzymes, cofactors
required for catalytic reactions
2. Cytochrome P450 oxidative system: Catalytic cycle of P450 reactions, mechanism of
P450 hydroxylation reactions, introduction to CYP450 superfamily of enzymes and their
classification, human CYP450s involved in drug metabolism and their typical substrates,
inhibitors and inducers.
3. Introduction to other drug metabolism enzyme isoforms/families
Glucuronosyltransferases, glutathione transferases, sulfotransferases, N-
acetyltransferases, FMO’s.
4. Methods for studying drug metabolism: Isolated enzymes, recombinant enzymes,
subcellular fractions, hepatocytes, perfused liver, in-vivo drug metabolism studies –
introduction to these methods, their utility, advantages and limitations
5. Introduction to in-silico methods for predicting drug metabolism: Principles behind
development of these systems, their potential and their limitations.

8. Environmental biotechnology
1. Application of biotechnology in agriculture e.g. pest control, herbicide resistance
2. GM crops and farm animals, bio-fertilizers
3. Alternative energy resources including biogas, alcohol etc.
4. Treatment of waste from domestic, industrial, agricultural etc. and bioremediation
Environmental security and safety; Socio-economic aspects of GM crops

9. Intellectual property Rights and Patent Filing


1. Introduction to IP
2. Copyright, Related Rights, Trademarks, Geographical Indications, Industrial Design
3. Patents
4. WIPO Treaties
5. Unfair Competition
6. Protection of New Varieties of Plants
7. Summary and Discussion on IP Rights

10. Molecular Biology


1. Introduction to recombinant DNA technology: Introduction to DNA
and its functions, Replication of DNA and its transcription and translation, restriction
enzymes and their properties, vectors for use in rDNA technology, creation and
introduction of rDNA molecules, cloning and expression of rDNA molecules, cloning
and expression systems, their advantages and limitations, application of rDNA
technology in production of pharmaceutical and in drug discovery and development.
2. High throughput screening: Introduction to the principles of screening and the philosophy
of HTS, considerations in HTS method development, validation of HTS methodology,
some examples of typical HTS assays and the principles involved therein.
3. Human Genome Initiative: Introduction to the genome, genome complexity and genome
organization, basic approaches towards sequencing of genomes, the approach for
sequencing the human genome, sources for obtaining human genome sequence
information, data mining of the human genome sequence for information and other
potential applications, introduction to bioinformatics.

11. Pharmacology, Toxicology and Therapeutics


1. Evaluation of drug activities,
2. Study models for testing,
3. Toxicity (ICH and OECD guidelines),
4. Importance of transgenic animal models/knock out mice based screening methods,
overview of regulatory status-ethical/moral/professional issues in toxicity

12. Process Biotechnology


1. Selection of separation process. Chemical, physical and biochemical aspects of isolation
and purification of biomolecules. Product release from a cell
2. Concentration and separation methods: membrane, ion-exchange, precipitation and
extraction. Chromatographic methods of purification
3. Chemistry of adsorption, Adsorbents, Equilibria, Yield and purity, Batch adsorption,
Kinetic analysis, Discrete stage analysis, Adsorption in fixed beds
4. Design and scale-up of adsorption and chromatography equipment
5. Design of downstream processing equipment. Downstream process economics

13. Tissue Engineering and Biopolymers


1. Principles of materials science and cell biology underlying the design of medical implants,
artificial organs, and matrices for tissue engineering
2. Methods for biomaterials surface characterization and analysis of protein adsorption on
biomaterials
3. Molecular and cellular interactions with biomaterials are analyzed in terms of unit cell
processes, such as matrix synthesis, degradation, and contraction Mechanisms underlying
wound healing and tissue remodeling following implantation in various organs
4. Tissue and organ regeneration
5. Design of implants and prostheses based on control of biomaterials-tissue interactions
6. Comparative analysis of intact, biodegradable, and bioreplaceable implants by reference to
case studies
7. Criteria for restoration of physiological function for tissues and organs

14. Targeted Drug Delivery Systems


1. Introduction: concept, basis, need, physicochemical and physiological basis, RES
2. Receptor mediated drug targeting
3. Colon targeting approaches
4. Targeting to brain
5. Targeting in cancer and infectious diseases
6. Ligands for targeted drug delivery: monoclonal antibodies

15.Unit Operation in Bioprocesses


1. Downstream Processing in Biotechnology, Selection of unit operation with due
consideration of physical, chemical and biochemical aspect of biomolecules, basic review of
bioprocess designing.
2. Primary separation and recovery processes: Cell disruption methods for intracellular
products, removal of insolubles, biomass (and particulate debris) separation techniques,
flocculation and sedimentation, centrifugation and filtration methods.
3. Enrichment operations: Membrane – based separations (micro and ultrafiltration,
precipitation methods, extractive separation, aqueous two-phase extraction, supercritical
extraction, insitu product removal, integrated bioprocessing.
4. Product resolution / fractionation: Adsorptive chromatographic separations processes,
electrophoretic separations, hybrid separation technologies (electrochromatography).
5. Product finishing: precipitation/crystallization, mixing, dialysis, distillation and drying.
Ultracentrifugation as a separation technique for fractionation of cells and proteins.
6. Introduction to Process Analytical Technology (PAT) and Quality by Design (QbD). Scale
down, monitoring and Validation of bioprocesses

16. Environmental Biotechnology


1. Environmental impact and control; Biosafety
2. Biological treatment: stabilization pond, aerated lagoon, activated sludge process, trickling
filter anaerobic treatment
3. Biodegradation of xenobiotic organic chemicals; Biological Detoxification of Hazardous
chemicals
4. Environmental Policy & Legislation; Sampling of air and water pollutants; Monitoring
techniques and methodology, pH, Dissolved Oxygen (DO); Chemical oxygen demand
(COD); Biological Oxygen Demand (BOD); Speculation of metals, monitoring & analysis
of CO, NO2, CO2, SO2; Pesticide residue; Phenols and petrochemicals
5. Environmental pollution control- Bioremediation, Bioaugmentation and Biostimulation;
Biofilms in treatment of waste water; Biofilm development and biofilm Kinetics; Aerobic
Biofilms; Bioreactors for waste water treatments

17. Biocatalysis and Green Technology


1. Catalytic activity of biomolecules – enzymes and ribozymes; Enzyme applications:
Hydrolase enzymes – lipases, esterases, proteases etc. with specific examples and
mechanism, Lyases – e.g. Aspartase, tyrosine-phenol lyase; Isomerases – e.g. glucose
isomerise; Transferases – e.g. aminotransferases, PLP as cofactor; Ligases; Oxidoreductases
– dehydrogenases, oxidases, oxygenases, peroxidases.
2. Whole cells as catalysts; Energetically unfavourable reactions at low temperatures and in
unfavourable solvents; The Michaelis-Menten model and modes of inhibition; Kinetics of
enzyme catalysed reaction; Regulation mechanisms; Mechanisn of enzyme action; Multi-
enzyme systems; Selection and screening of biocatalysts for activity, stability and substrate
or product selectivity; Extremozymes – protein catalysts for reactions at extremes of
temperature, pressure and pH.
3. Principles of green chemistry (e.g. prevention of waste, less hazardous methods, safer
chemicals and solvents, energy efficiency, atom economy, use of catalysis, etc.); the design
of “greener” effect chemicals, with examples from the development of crop protection
agents; the design of “greener” chemical processes, with examples of the use of biocatalysts.

18. Carbohydrate chemistry and technology


1. Different carbohydrates in food products such as starch, cellulose, sugars, pectin, fibre etc.
and their significance in diet
2. Their chemistry & changes in them during processing; Chemical & enzymic modification;
Interactions with other food constituents and their implications
3. Special application of carbohydrates in gels, emulsions, stabilisation of food systems,
simulated and low-fat foods, edible packages etc.

19. Nutritional genomics


1. Gene- environment interaction; gene- diet interaction; principals and practice behind dietary
management of genetically transmitted disorders 10
2. Phenylketonuria, galactosemia; G6PD deficiency; lactose intolerance; complex traits; birth
disorders; signal transduction; epigenetic mechanism 10
3. Bioactive components of food; nutraceutical; effective gene expression; epigenetic process;
signal transduction. Recent developments in the field

IV. Laboratory courses

Instrumental methods of analysis


UV/Visible Spectroscopy
i. Calibration of UV spectrophotometer
ii. Study effect of solvent on wavelength maxima of drugs.
iii. Find Beer’s law limit of drugs in a suitable solvent.
iv. Standard calibration curve by UV spectroscopy at
a) λ max
b) λ max + 10 nm
c) λ max – 10 nm
v. Determination of pKa by U.V. spectroscopy.
vi. Multicomponent analysis by UV-Spectrophotometry
vii. Absorbance corrected for interference method
viii. Simultaneous equation method
ix. Absorbance ratio method
x. Area under curve method
xi. First derivative spectrophotometric method
Analysis of drugs from formulations focusing on separation of drug from the formulation
excipients
IR Spectroscopy
i. Calibration of IR spectrophotometer
ii. Sample preparation for I.R. spectroscopy (solid/liquids) and interpretation of IR bands for
important functional groups.
DSC analysis of drugs in crystalline and amorphous forms.
Chromatography:
i. HPLC calibration of HPLC column and determination of response factor by HPLC
ii. GC Instrumental handling and few analyses of the API intermediates
iii. TLC mobile phase selection of a various combination of compounds and reaction
monitoring.
iv. Preparative TLC analysis.
v. pH stability evaluation of a drug by TLC.
vi. Separation of components by column chromatography.
Structural Interpretation by Spectroscopy:
i. Basic interpretations of simple Mass spectra and NMR.
ii. Structural elucidation workshop: Interpretation of 1H NMR, 13C NMR, IR and Mass
spectrometry of simple compounds (maximum 12 carbon atoms).

Microbiology & Molecular Biology Laboratory


1. Study of bacteria, yeasts, moulds, algae, viruses and other microorganisms
2. Morphology, structure, reproduction, isolation, and cultivation
3. Principles of taxonomy and classification, Mutants, Control of microorganisms
4. Laboratory experiments in use of microscopy for identification of microorganisms by
morphology and staining technique. Isolation of pure culture
5. Study of growth and optimisation of conditions
6. Preparation of culture media, Sterility test
7. Basic methods in Molecular Biology, including PCR, Blotting techniques, DNA
purification, DNA sequencing etc

You might also like