Personalized Medicine - Scientific & Commercial Aspects
NEW YORK, Nov. 30, 2011 /PRNewswire/ -- Reportlinker.com announces that a new market research report is available in its catalogue:
Personalized Medicine - scientific & commercial aspects
http://www.reportlinker.com/p0203549/Personalized-Medicine---scientific--commercial-aspects.html#utm_source=prnewswire&utm_medium=pr&utm_campaign=Genomics
Summary
The aim of personalized medicine or individualized treatment is to match the right drug to the right patient and, in some cases, even to design the appropriate treatment for a patient according to his/her genotype. This report describes the latest concepts of development of personalized medicine based on pharmacogenomics, pharmacogenetics,pharmacoproteomics, and metabolomics. Basic technologies of molecular diagnostics play an important role, particularly those for single nucleotide polymorphism (SNP) genotyping. Diagnosis is integrated with therapy for selection of the treatment as well for monitoring the results. Biochip/microarray technologies are also important and finally bioinformatics is needed to analyze the immense amount of data generated by various technologies.
Pharmacogenetics, the study of influence of genetic factors on drug action and metabolism, is used for predicting adverse reactions of drugs. Several enzymes are involved in drug metabolism of which the most important ones are those belonging to the family of cytochrome P450. The knowledge of the effects of polymorphisms of genes for the enzymes is applied in drug discovery and development as well as in clinical use of drugs. Cost-effective methods for genotyping are being developed and it would be desirable to include this information in the patient's record for the guidance of the physician to individualize the treatment. Pharmacogenomics, a term that overlaps with pharmacogenetics but is distinct, deals with the application of genomics to drug discovery and development. It involves the mechanism of action of drugs on cells as revealed by gene expression patterns. Pharmacoproteomics is an important contribution to personalized medicine as it is a more functional representation of patient-to-patient variation than that provided by genotyping.A 'pharmacometabonomic' approach to personalizing drug treatment is also described.
Biological therapies such as those which use patient's own cells are considered to be personalized medicines. Vaccines are prepared from individual patient's tumor cells. Individualized therapeutic strategies using monoclonal bodies can be directed at specific genetic and immunologic targets. Ex vivo gene therapy involves the genetic modification of the patient's cells in vitro, prior to reimplantation of these cells in the patient's body.
Various technologies are integrated to develop personalized therapies for specific therapeutic areas described in the report. Examples of this are genotyping for drug resistance in HIV infection, personalized therapy of cancer, antipsychotics for schizophrenia, antidepressant therapy, antihypertensive therapy and personalized approach to neurological disorders. Although genotyping is not yet a part of clinically accepted routine, it is expected to have this status by the year 2014.
Several players are involved in the development of personalized therapy. Pharmaceutical and biotechnology companies have taken a leading role in this venture in keeping with their future role as healthcare enterprises rather than mere developers of technologies and manufacturers of medicines.
Ethical issues are involved in the development of personalized medicine mainly in the area of genetic testing. These along with social issues and consideration of race in the development of personalized medicine are discussed. Regulatory issues are discussed mainly with reference to the FDA guidelines on pharmacogenomics.
Increase in efficacy and safety of treatment by individualizing it has benefits in financial terms. Information is presented to show that personalized medicine will be cost-effective in healthcare systems. For the pharmaceutical companies, segmentation of the market may not leave room for conventional blockbusters but smaller and exclusive markets for personalized medicines would be profitable. Marketing opportunities for such a system are described with market estimates from 2010-2020.
Profiles of 245 companies involved in developing technologies for personalized medicines, along with 459 collaborations are included in the part II of the report. Finally the bibliography contains over 600 selected publications cited in the report.The report is supplemented by 63 tables and 17 figures.
TABLE OF CONTENTS
0. Executive Summary 19
1. Basic Aspects 21
Definition of personalized medicine 21
History of medical concepts relevant to personalized medicine 22
Molecular biological basis of personalized medicine 24
The human genome 24
Chromosomes 25
Genes 25
The genetic code 25
Gene expression 25
DNA sequences and structure 26
Genetic variations in the human genome 26
Single nucleotide polymorphisms 27
Copy number variations in the human genome 27
Insertions and deletions in the human genome 29
Large scale variation in human genome 29
Structural variations in the human genome 30
Mapping and sequencing of structural variation from human genomes 30
1000 Genomes Project 31
Role of DNA sequencing in the development of personalized medicine 32
Human Variome Project 33
Interconnected genetic and genomic patterns in human diseases 33
Basics technologies for developing personalized medicine 34
Definitions of technologies relevant to personalized medicine 34
Problems with the ICH definitions of pharmacogenomcis and pharmacogenetics 34
Relationship of various technologies to personalized medicine 35
Conventional medicine versus personalized medicine 35
Role of genetics in future approaches to healthcare 36
Genetic medicine 36
Human disease and genes 36
Genetic and environmental interactions in etiology of human diseases 37
Role of genetics in development of personalized medicines 37
Genetic databases 37
Genetic epidemiology 38
Limitations of medical genetics and future prospects 38
Genetics vs. epigenetics 39
Role of systems biology in personalized medicine 39
Systems pharmacology 40
Systems medicine 41
Synthetic biology and development of personalized medicines 41
A personalized approach to environmental factors in disease 42
Reclassification of diseases 43
2. Molecular Diagnostics in Personalized Medicine 45
Introduction 45
Molecular diagnostic technologies 45
PCR-based methods 46
DirectLinear™ Analysis 46
Denaturing high-performance liquid chromatography 47
Multiplex Allele-Specific Diagnostic Assay 47
Representational oligonucleotide microarray analysis 47
Restriction fragment length polymorphism (RFLP) 47
Real-time PCR for detection of CNVs 47
Non-PCR methods 48
Arrayed primer extension (APEX) 48
Enzymatic Mutation Detection (EMD) 48
DNA sequencing 48
Sanger-sequencing technology 49
ABI PRISM® 310 Genetic Analyzer 50
High-throughput paired end transcriptome sequencing 50
Emerging sequencing technologies 50
4300 DNA analyzer 51
Apollo 100 51
"Color blind" approach to DNA sequencing 52
Cyclic array sequencing 52
CEQ™ 8000 52
DeepCAGE sequencing 52
Electron microscope-based DNA sequencing 53
Genometrica? sequencer 53
GS-FLEX system (Roche/454) 54
IBS sequencing technology 55
Illumina Genome Analyzer System 55
MegaBACE 500 56
Microdroplet-based PCR for large-scale targeted sequencing. 56
Multiplex amplification of human DNA sequences 57
Nanoscale sequencing 57
Polonator sequencer 57
RainStorm™ microdroplet technology 58
Sequential DEXAS 58
SOLiD technology 59
Sequencing by hybridization 60
Whole genome sequencing 60
Bioinformatic tools for analysis of genomic sequencing data 60
Detection of single molecules in real time 61
Direct observation of nucleotide incorporation 61
Molecular Combing 61
Nanopore sequencing 61
DNA sequence by use of nanoparticles 62
Zero-mode waveguide nanostructure arrays 62
Future prospects of sequencing 62
Role of sequencing in development of personalized medicine 63
Biochips and microarrays 63
Application of biochip technology in developing personalized medicine 63
Standardizing the microarrays 65
Biochip technologies 65
GeneChip 65
AmpliChip CYP450 65
Microfluidics 67
Lab-on-a-chip 67
Micronics' microfluidic technology 68
LabCD 68
Microfluidic automated DNA analysis using PCR 68
Integrated microfluidic bioassay chip 68
Electronic detection of nucleic acids on microarrays 69
Strand displacement amplification on a biochip 69
Rolling circle amplification on DNA microarrays 70
Universal DNA microarray combining PCR and ligase detection reaction 70
Protein biochips 70
ProteinChip 71
LabChip for protein analysis 72
TRINECTIN proteome chip 72
Protein expression microarrays 72
Microfluidic devices for proteomics-based diagnostics 73
New developments in protein biochips/microarrays 73
Protein biochips/microarrays for personalized medicine 74
SNP genotyping 74
Genotyping and haplotyping 75
Haplotype Specific Extraction 76
Computation of haplotypes 76
HapMap project 76
Haplotyping for whole genome sequencing 78
Predictingdrug response with HapMap 78
Companies developing haplotyping technology 78
Technologies for SNP analysis 79
Biochip and microarray-based detection of SNPs 80
SNP genotyping by MassARRAY 80
Biochip combining BeadArray and ZipCode technologies 80
SNP-IT primer-extension technology 80
Affymetrix Variation Detection Arrays 81
Use of NanoChip for detection of SNPs 81
Electrochemical DNA probes 81
Single base extension-tag array 82
Laboratory Multiple Analyte Profile 82
PCR-CTPP (confronting two-pair primers) 83
SNP genotyping on a genome-wide amplified DOP-PCR template 83
TaqMan real-time PCR 83
Non-Enzymatic Amplification Technology 83
SNP genotyping with gold nanoparticle probes 84
Locked nucleic acid 84
Molecular inversion probe based assays 84
Pyrosequencing 85
Reversed enzyme activity DNA interrogation test 85
Smart amplification process version 2 86
Zinc finger proteins 86
UCAN method (Takara Biomedical) 86
Mitochondrial SNPs 87
Limitations of SNP in genetic testing 87
Concluding remarks on SNP genotyping 87
Companies involved in developing technologies/products for SNP analysis 88
Impact of SNPs on personalized medicine 89
Detection of copy number variations 90
Study of rare variants in pinpointing disease-causing genes 90
Optical Mapping 91
Role of nanobiotechnology in molecular diagnostics 91
Cantilevers for personalized medical diagnostics 92
Nanopore-based technology for single molecule identification 92
Role of biomarkers in personalized medicine 93
Biomarkers for diagnostics 93
Biomarkers for drug development 94
Application of proteomics in molecular diagnosis 94
Proteomic strategies for biomarker identification 94
Proteomic technologies for detection of biomarkers in body fluids 94
Protein patterns 95
Layered Gene Scanning 95
Comparison of proteomic and genomic approaches in personalized medicine 96
Gene expression profiling 96
DNA microarrays 97
Analysis of single-cell gene expression 97
Gene expression profiling based on alternative RNA splicing 98
Whole genome expression array 99
Tangerine™ expression profiling 99
Gene expression analysis on biopsy samples 100
Profiling gene expression patterns of white blood cells 100
Serial analysis of gene expression (SAGE) 101
Multiplexed Molecular Profiling 101
Gene expression analysis using competitive PCR and MALDI TOF MS 102
Monitoring in vivo gene expression by magnetic resonance imaging 102
Companies involved in gene expression analysis 102
Monitoring in vivo gene expression by molecular imaging 103
Molecular imaging and personalized medicine 104
Glycomics-based diagnostics 104
Combination of diagnostics and therapeutics 104
Use of molecular diagnostics for stratification in clinical trials 105
Companion diagnostics 105
Companies involved in companion diagnostics 105
Point-of-care diagnosis 107
Companies developing point-of-care diagnostic technologies 108
Point-of-care diagnosis of infections 110
Advantages versus disadvantages of point-of-care diagnosis 111
Future prospects of point-of-care diagnosis 111
Genetic testing for disease predisposition 112
Preventive genetics by early diagnosis of mitochondrial diseases 112
Direct-to-consumer genetic services 112
Role of diagnostics in integrated healthcare 114
Concept of integrated healthcare 114
Components of integrated healthcare 115
Screening 115
Disease prediction 115
Early diagnosis 115
Prevention 115
Therapy based on molecular diagnosis 115
Monitoring of therapy 115
Advantages and limitations of integrated healthcare 116
Commercially available systems for integrated healthcare 116
Future of molecular diagnostics in personalized medicine 117
3. Pharmacogenetics 119
Basics of pharmacogenetics 119
Role of molecular diagnostics in pharmacogenetics 120
Role of pharmacogenetics in pharmaceutical industry 121
Study of the drug metabolism and pharmacological effects 121
Causes of variations in drug metabolism 121
Enzymes relevant to drug metabolism 122
Pharmacogenetics of phase I metabolism 122
CYP450 122
P450 CYP 2D6 inhibition by selective serotonin reuptake inhibitors 124
Cytochrome P450 polymorphisms and response to clopidogrel 125
Lansoprazole and cytochrome P450 125
Glucose-6-phosphate dehydrogenase 125
Pharmacogenetics of phase II metabolism 126
N-Acetyltransferase 126
Uridine diphosphate-glucuronosyltransferase 127
Measurement of CYP isoforms 127
Polymorphism of drug transporters 128
Genetic variation in drug targets 128
Polymorphisms of kinase genes 129
Effect of genetic polymorphisms on disease response to drugs 129
Ethnic differences in drug metabolism 130
Gender differences in pharmacogenetics 130
Role of pharmacogenetics in drug safety 131
Adverse drug reactions 131
Adverse drug reactions in children 132
Adverse drug reactions related to toxicity of chemotherapy 132
Genetically determined adverse drug reactions 132
Malignant hyperthermia 134
Pharmacogenetics of clozapine-induced agranulocytosis 134
Role of pharmacogenetics in antiplatelet therapy with clopidrogrel 134
Role of pharmacogenetics in warfarin therapy 135
Role of pharmacogenetics in antiplatelet therapy 136
Role of pharmacogenetics in carbamazepine therapy 137
Role of pharmacogenetics in statin therapy 137
FDA consortium linking genetic biomarkers to serious adverse events 137
Therapeutic drug monitoring, phenotyping, and genotyping 138
Therapeutic drug monitoring 138
Phenotyping 139
Genotyping 140
Genotyping vs phenotyping 140
Phenomics 141
Limitations of genotype-phenotype association studies 141
Molecular toxicology in relation to personalized medicines 142
Toxicogenomics 142
Biomarkers of drug toxicity 142
Drug-induced mitochondrial toxicity 143
Companies involved in molecular toxicology 143
Gene expression studies 144
Pharmacogenetics in clinical trials 144
Postmarketing pharmacogenetics 145
Clinical implications of pharmacogenetics 145
Application of CYP450 genotyping in clinical practice 145
Pharmacogenomic biomarker information in drug labels 145
Genotype-based drug dose adjustment 145
Examples of use of pharmacogenetics in clinical pharmacology 146
Genotyping for identifying responders to sulfasalazine 146
HLA alleles associated with lumiracoxib-related liver injury 146
Pharmacogenetic basis of thiopurine toxicity 147
Tranilast-induced hyperbilirubinemia due to gene polymorphism 147
Linking pharmacogenetics with pharmacovigilance 147
Genetic susceptibility to ADRs 147
Linking genetic testing to postmarketing ADR surveillance 147
Recommendations for the clinical use of pharmacogenetics 148
Limitations of pharmacogenetics 148
Pharmacoepigenomics vs pharmacogenetics in drug safety 149
Future role of pharmacogenetics in personalized medicine 149
4. Pharmacogenomics 151
Introduction 151
Basics of pharmacogenomics 152
Pharmacogenomics and drug discovery 152
Preclinical prediction of drug efficacy 154
Pharmacogenomics and clinical trials 154
Impact of genetic profiling on clinical studies 155
Limitations of the pharmacogenomic-based clinical trials 156
Pharmacogenomic aspects of major therapeutic areas 157
Oncogenomics 157
Oncogenes 157
Tumor suppressor genes 158
Cardiogenomics 159
Neuropharmacogenomics 161
Pharmacogenomics of Alzheimer's disease 161
Pharmacogenomics of depression 162
Pharmacogenomics of schizophrenia 162
Companies involved in neurogenomics-based drug discovery 163
5. Role of Pharmacoproteomics 165
Basics of proteomics 165
Proteomic approaches to the study of pathophysiology of diseases 165
Single cell proteomics for personalized medicine 166
Diseases due to misfolding of proteins 166
Therapies for protein misfolding 167
Significance of mitochondrial proteome in human disease 168
Proteomic technologies for drug discovery and development 168
Role of reverse-phase protein microarray in drug discovery 168
Role of proteomics in clinical drug safety 168
Toxicoproteomics 169
Application of pharmacoproteomics in personalized medicine 170
6. Role of Metabolomics in Personalized Medicine 171
Metabolomics and metabonomics 171
Metabolomics bridges the gap between genotype and phenotype 171
Metabolomics, biomarkers and personalized medicine 172
Metabolomic technologies 172
Urinary profiling by capillary electrophoresis 173
Lipid profiling 173
Role of metabolomics in biomarker identification and pattern recognition 174
Validation of biomarkers in large-scale human metabolomics studies 174
Pharmacometabonomics 174
Metabonomic technologies for toxicology studies 175
Metabonomics/metabolomics and personalized nutrition 175
7. Personalized Biological Therapies 177
Introduction 177
Recombinant human proteins 177
Therapeutic monoclonal antibodies 177
Cell therapy 178
Autologous tissue and cell transplants 178
Stem cells 178
Role of stem cells derived from unfertilized embryos 178
Cloning and personalized cell therapy 179
Use of stem cells for drug testing 179
Gene therapy 179
Personalized vaccines 180
Personalized vaccines for viral diseases 180
Personalized cancer vaccines 180
Antisense therapy 180
RNA interference 181
MicroRNAs 182
8. Personalized Medicine in Major Therapeutic Areas 183
Introduction 183
Management of infections 184
Management of HIV 184
CD4 counts as a guide to drug therapy for AIDS 184
Drug-resistance in HIV 184
Genetics of human susceptibility to HIV infection 185
Measurement of Replication Capacity 186
Personalized vaccine for HIV 186
Prevention of adverse reactions to antiviral drugs 186
Pharmacogenetics and HIV drug safety 187
Pharmacogenomics of antiretroviral agents 187
Role of diagnostic testing in HIV 188
Role of genetic variations in susceptibility to HIV-1 188
Personalized treatment of hepatitis B 188
Personalized treatment of hepatitis C 189
Personalized management of tuberculosis 190
Psychiatric disorders 191
Psychopharmacogenetics 191
COMT genotype and response to amphetamine 191
Genotype and response to methylphenidate in children with ADHD 192
Personalized antipsychotic therapy 192
Personalized antidepressant therapy 194
EEG to predict adverse effects and evaluate antidepressant efficacy 195
Individualization of SSRI treatment 195
Vilazodone with a test for personalized treatment of depression 197
Neurological disorders 197
Personalized management of Alzheimer's disease 197
Personalized management of Parkinson's disease 198
Discovery of subgroup-selective drug targets in PD 199
Personalized management of Epilepsy 199
Choice of the right AED 200
Pharmacogenetics of epilepsy 200
Pharmacogenomics of epilepsy 200
Drug resistance in epilepsy 201
Future prospects for management of epilepsy 202
Personalized management of migraine 203
Individualization of use of triptans for migraine 203
Personalized management of stroke 204
Brain imaging in trials of restorative therapies for stroke 204
Decisions for evacuation of intracerebral hemorrhage 204
Revascularization procedures in chronic post-stroke stage 205
Personalized treatment of multiple sclerosis 205
Immunopathological patterns of demyelination for assessing therapy 206
Personalizing mitoxantrone therapy of multiple sclerosis 206
Fusokine method of personalized cell therapy of multiple sclerosis 207
MBP8298 207
Pharmacogenomics of IFN-? therapy in multiple sclerosis 207
T cell-based personalized vaccine for MS 209
Cardiovascular disorders 209
Role of diagnostics in personalized management of cardiovascular disease 209
Testing in coronary heart disease 209
SNP genotyping in cardiovascular disorders 210
Cardiovascular disorders with a genetic component 210
Gene variant as a risk factor for sudden cardiac death 212
KIF6 gene test as a guide to management of heart disease 212
SNP Chip for study of cardiovascular diseases 213
Pharmacogenomics of cardiovascular disorders 213
Modifying the genetic risk for myocardial infarction 213
Management of heart failure 214
?-blockers 214
Bucindolol 214
BiDil 214
Management of hypertension 215
Pharmacogenomics of diuretic drugs 215
Pharmacogenomics of ACE inhibitors 216
Management of hypertension by personalized approach 216
Prediction of antihypertensive activity of rostafuroxin 217
Pharmacogenetics of lipid-lowering therapies 218
Polymorphisms in genes involved in cholesterol metabolism 218
Role of eNOS gene polymorphisms 219
The STRENGTH study 219
Personalized management of women with hyperlipidemia 220
Thrombotic disorders 221
Factor V Leiden mutation 221
Anticoagulant therapy 221
Antiplatelet therapy 222
Nanotechnology-based personalized therapy of cardiovascular diseases 222
Project euHeart for personalized management of heart disease 223
Concluding remarks 223
Personalized management of pulmonary disorders 223
Role of genetic ancestory in lung function 224
Personalized therapy of asthma 224
Biomarkers for predicting response to corticosteroid therapy 224
Genetic polymorphism and response to ?2-adrenergic agonists 225
Genotyping in asthma 225
IgE as guide to dosing of omalizumab for asthma 226
Lebrikizumab for personalised treatment of asthma 226
Personalized management of chronic obstructive pulmonary disease 226
Personalized management of skin disorders 227
Genetic testing for personalized skin care 227
Management of hair loss based on genetic testing 228
Personalized therapy of rheumatoid arthritis 228
DIATSTAT™ anti-cyclic citrullinated peptides in rheumatoid arthritis 229
Personalization of COX-2 inhibitor therapy 229
Personalization of infliximab therapy 230
Personalized approaches in immunology 230
Role of Mannose-binding lectin in personalized medicine 231
Pharmacogenetics and pharmacogenomics of immunosuppressive agents 231
Personalized management of patients with lupus erythematosus 231
Personalized management of pain 232
Pharmacogenetics/pharmacogenomics of pain 232
Mechanism-specific management of pain 233
Preoperative testing to tailor postoperative analgesic requirements 233
Personalized analgesics 234
Management of genetic disorders 234
Personalized treatment of cystic fibrosis 234
Personalized management of gastrointestinal disorders 235
Personalized therapy of inflammatory bowel disease 235
Personalized management of lactose intolerance 236
Personalized approaches to improve organ transplantation 236
Personalization of kidney transplantation 236
Personalization of cardiac transplantation 237
Prediction of rejection to tailor anti-rejection medications 237
Personalized immunosuppressant therapy in organ transplants 238
Role of immunological biomarkers in monitoring grafted patients 239
Improved matching of blood transfusion 239
Personalized approach to addiction 239
Pharmacogenetics of drug addiction 239
Genetic polymorphism and management of alcoholism 240
Personalized therapy for smoking cessation 240
Antidepressant therapy for smoking cessation 241
Effectiveness of nicotine patches in relation to genotype 241
Personalized approaches to miscellaneous problems 241
Hormone replacement therapy in women 241
Personalized treatment of malaria 242
Personalized management of renal disease 242
Gene associated with end-stage renal disease 243
Personalized care of trauma patients 243
Personalized anticoagulation 244
Personalized Hyperbaric oxygen therapy 244
Personalized preventive medicine 245
Personalized nutrition 245
Nutrigenomics 246
Genomics of vitamin D and calcium supplementation 246
Nutrigenomics and functional foods 247
Nutrigenetics and personalized medicine 247
Nutrigenomics and personalized medicine248
Nutrition and proteomics 248
Personalized diet prescription 248
9. Personalized Therapy of Cancer 251
Introduction 251
Challenges of cancer classification 251
Relationships of technologies for personalized management of cancer 251
Impact of molecular diagnostics on the management of cancer 252
AmpliChip P53 as companion diagnostic for cancer 253
Analysis of RNA splicing events in cancer 253
Analysis of chromosomal alterations in cancer cells 253
Cancer classification using microarrays 253
Detection of loss of heterozygosity 254
Diagnosis of cancer of an unknown primary 255
Diagnostics for detection of minimal residual disease 255
DNA repair biomarkers 255
Fluorescent in situ hybridization 256
Gene expression profiling 256
Gene expression profiles predict chromosomal instability in tumors 257
Isolation and characterization of circulating tumor cells 258
Modulation of CYP450 activity for cancer therapy 258
Personalized therapies based on oncogenic pathways signatures 259
Quantum dot-based test for DNA methylation 259
Role of molecular imaging in personalized therapy of cancer 259
Functional diffusion MRI 260
FDG-PET/CT for personalizing cancer treatment 260
Image-guided personalized drug delivery in cancer 261
Tumor imaging and elimination by targeted gallium corrole 261
Future prospects of molecular imaging in management of cancer 261
Unraveling the genetic code of cancer 262
Cancer prognosis 262
Detection of mutations for risk assessment and prevention 263
Impact of biomarkers on management of cancer 263
HER-2/neu oncogene as a biomarker for cancer 263
L-asparaginase treatment of cancer guided by a biomarker 264
Oncogene GOLPH3 as a cancer biomarker 264
Predictive biomarkers for cancer 264
Sequencing to discover biomarkers to personalize cancer treatment 265
Systems biology approach to discovery of radiation sensitivity biomarkers 265
VeraTag™ assay system for cancer biomarkers 266
Determination of response to therapy 266
ChemoFx cell culture assay for predicting anticancer drug response 266
Ex vivo testing of tumor biopsy for chemotherapy sensitivity 267
Genomic approaches to predict response to anticancer agents 267
Gene expression patterns to predict response of cancer to therapy 267
Genomic analysis of tumor biopsies 268
Genotype-dependent efficacy of pathway inhibition in cancer 268
Mutation detection at molecular level 268
Role of genetic variations in susceptibility to anticancer drugs 269
Non-genetic factors for variations in response of cancer cells to drugs 269
Proteomic analysis of tumor biopsies to predict response to treatment 269
Real-time apoptosis monitoring 270
Serum nucleosomes as indicators of sensitivity to chemotherapy 270
Targeted microbubbles to tumors for monitoring anticancer therapy 271
PET imaging for determining response to chemotherapy 271
Tissue systems biology approach to personalized management of cancer 271
Targeted cancer therapies 272
Targeting glycoproteins on cell surface 272
Targeting pathways in cancer 272
Functional antibody-based therapies 272
Personalized cancer vaccines 274
Antigen-specific vaccines 274
Active immunotherapy based on antigen specific to the tumor 274
Tumor-derived vaccines 275
MyVax 275
OncoVAX 275
Tumor cells treated with dinitrophenyl 276
Prophage 276
Melacine 276
Patient-specific cell-based vaccines 277
Dendritic cell-based vaccines 277
Adoptive cell therapy 278
Combination of antiangiogenic agents with ACT 280
Genetically targeted T cells for treating B cell malignancies 280
Genetic engineering of tumor cells 280
Hybrid cell vaccination 281
Personalized peptide cancer vaccines 281
Current status and future prospects of personalized cancer vaccines 281
Personalized radiation therapy 283
Molecular diagnostics combined with cancer therapeutics 284
Aptamers for combined diagnosis and therapeutics of cancer 284
Role of nanobiotechnology in personalized management of cancer 285
Design of future cancer therapies 286
Screening for personalized anticancer drugs 286
Role of epigenetics in development of personalized cancer therapies 286
Personalized therapy of cancer based on cancer stem cells 287
Role of oncoproteomics in personalized therapy of cancer 287
Cancer tissue proteomics 287
Role of sequencing in personalized therapy of cancer 288
Pharmacogenomic-based chemotherapy 288
Whole genome technology to predict drug resistance 288
Anticancer drug selection based on molecular characteristics of tumor 288
Testing microsatellite-instability for response to chemotherapy 289
Pharmacogenetics of cancer chemotherapy 289
CYP 1A2 290
Thiopurine methyltransferase 290
Dihydropyrimidine dehydrogenase 291
UGT1A1 test as guide to irinotecan therapy 291
Role of computational models in personalized anticancer therapy 292
A computational model of kinetically tailored treatment 292
Mathematical modeling of tumor mivroenvironments 292
Molecular profiling of cancer 293
Drug resistance in cancer 293
Detection of drug resistance in cancer by metabolic profiling 294
Determination of chemotherapy response by topoisomerase levels 294
Anaplastic lymphoma kinase 295
Management of drug resistance in leukemia 295
Overexpression of multidrug resistance gene 295
P53 mutations 296
A chemogenomic approach to drug resistance 296
Systems biology approach to personalizing therapy for drug-resistant cancer 296
Examples of personalized management of cancer 297
Personalized management of brain cancer 297
Biosimulation approach to personalizing treatment of brain cancer 297
Genetics and genomics of brain cancer 297
Prognosis of glioblastoma multiforme based on its genetic landscape 299
Molecular diagnostics for personalized management of brain cancer 299
Personalized chemotherapy of brain tumors 301
Personalized therapy of oligodendroglial tumors (OTs) 302
Personalized therapy of neuroblastomas 303
Personalized therapy of medulloblastomas 303
Personalized management of germ cell brain tumors 304
Personalized management of breast cancer 304
Developing personalized drugs for breast cancer 304
Gene expression plus conventional predictors of breast cancer 305
Her2 testing in breast cancer as a guide to treatment 306
HER2/neu-derived peptide vaccine for breast cancer 308
Molecular diagnostics in breast cancer 308
Pharmacogenetics of breast cancer 310
Proteomics-based personalized management of breast cancer 310
Predicting response to chemotherapy in breast cancer 310
Prediction of resistance to chemotherapy in breast cancer 313
Prediction of adverse reaction to radiotherapy in breast cancer 314
Prediction of recurrence in breast cancer for personalizing therapy 314
Prognosistic tests for breast cancer 316
Racial factors in the management of breast cancer 318
RATHER consortium to study personalized approach to breast cancer 318
TAILORx (Trial Assigning Individualized Options for Treatment) 318
Trends and future prospects of breast cancer research 319
Understanding tumor diversity in mouse mammary cancer model 319
Personalized management of ovarian cancer 319
Early diagnosis of ovarian cancer 320
Determining response to chemotherapy in ovarian cancer 320
Recurrent and drug-resistant ovarian cancer 320
Pathway targeted therapies for ovarian cancer 321
Personalized management of hematological malignancies 322
Personalized management of acute lymphoblastic leukemia 322
Personalized management of acute myeloid leukemia 323
Personalized management of chronic lymphocytic leukemia 324
Personalized management of multiple myeloma 324
Personalized management B cell lymphomas 325
Personalized vaccine for follicular lymphoma 326
Personalized management of myelodysplastic syndrome 327
Personalized management of hepatocellular carcinoma 327
Personalized management of gastrointestinal cancer 327
Personalized management of esophageal cancer 327
Personalized management of gastric cancer 328
Personalized management of colorectal cancer 328
A systems biology approach to drug resistance in colorectal cancer 331
Personalized management of liver cancer 331
Personalized management of lung cancer 332
Determination of outcome of EGFR tyrosine kinase inhibitor treatment 332
Crizotinib for personalized management of NSCLC 334
Testing for response to chemotherapy in lung cancer 334
Testing for prognosis of lung cancer 335
Testing for recurrence of lung cancer 335
Role of a new classification system in the management of lung cancer 335
Personalized management of malignant melanoma 336
Therapy for inhibiting BRAF mutation in melanoma 336
Vaccine for malignant melanoma based on heat shock protein 336
Personalized management of pancreatic cancer 337
Biomarkers of pancreatic cancer 337
Histone modifications predict treatment response in pancreatic cancer 338
Personlized management of prostate cancer 338
Diagnostics for guiding therapy of prostate cancer 338
Early detection of cancer recurrence and guiding treatment 339
Effects of of lifestyle changes shown by gene expression studies 339
Personalized peptide vaccine for prostate cancer 340
Future of cancer therapy 340
Challenges for developing personalized cancer therapies 340
Cancer Genome Atlas 341
COLTHERES consortium 341
Computer and imaging technologies for personalizing cancer treatment 341
Genomic Cancer Care Alliance 342
Integrated genome-wide analysis of cancer for personalized therapy 342
International Cancer Genome Consortium 342
PREDICT Consortium 343
Companies involved in developing personalized cancer therapy 344
10. Development of Personalized Medicine 347
Introduction 347
Non-genomic factors in the development of personalized medicine 347
Personalized medicine based on circadian rhythms 347
Cytomics as a basis for personalized medicine 348
Intestinal microflora 348
Gut microbiome compared to human genome 348
Metabolic interactions of the host and the intestinal microflora 349
Role of drug delivery in personalized medicine 349
Personalized approach to clinical trials 350
Use of Bayesian approach in clinical trials 350
Individualzing risks and benefits in clinical trials 350
Clinical trials of therapeutics and companion diagnostics 351
Players in the development of personalized medicine 351
Personalized Medicine Coalition 351
European Personalized Medicine Diagnostics Association 352
Role of pharmaceutical industry 353
Production and distribution of personalized medicines 353
Role of biotechnology companies 354
Role of life sciences industries 354
Role of molecular imaging in personalized medicine 355
Molecular imaging for personalized drug development in oncology 355
Molecular imaging and CNS drug development 357
Companies involved in molecular imaging 358
Role of the clinical laboratories 358
Role of the US government in personalized medicine 359
Department of Health and Human Services and personalized medicine 360
Agency for Healthcare Research and Quality 360
Comparative effectiveness research 361
Role of the US Governmen
To order this report:
Genomics Industry: Personalized Medicine - scientific & commercial aspects
Check our Industry Analysis and Insights
CONTACT
Nicolas Bombourg
Reportlinker
Email: [email protected]
US: (805)652-2626
Intl: +1 805-652-2626
SOURCE Reportlinker
WANT YOUR COMPANY'S NEWS FEATURED ON PRNEWSWIRE.COM?
Newsrooms &
Influencers
Digital Media
Outlets
Journalists
Opted In
Share this article