You are on page 1of 11

Index

Note: Page numbers followed by f indicate figures, and t indicate tables.

A experimental and analytical variability


Absolute humidity (AH), 39 analytical method uncertainty, 199–200
Accelerated degradation tests (ADTs), 148 batch-batch variability, 204
Accelerated life test (ALT), 147–148 duration of exposure, 200
Accelerated predictive stability (APS), 71t, 411 replicate testing, 204
applications temperature and humidity control, 200–202
formulation process development, 8 unit-unit/sample-sample variability, 202–204
formulation prototype rank ordering, 8 failure of, 176
genotoxic degradation product, kinetic investigation of, 10 fixed humidity stability studies
initial retest period, 9–10 protocol design features, 60–62, 61t
initial use period/shelf life for clinical supply, 9–10 saturated salt solutions, use of, 59
in-use stability, 10 water activity determinations, 60
mass balance investigation, 10 historical context of, 3–4
packaging selection, 8–9 humidity- and temperature-sensitivity coefficients, 68–70,
postapproval changes, assessment of, 11–12, 11t 68–69f, 70–71t
product physical changes, prediction of, 6 impact assessment, 232
registration, 10 initial marketing registration, 236–237
salt forms and polymorphs, comparative assessment isoconversional design, 152–153
of, 7–8 packaging selection, 232
small-molecule drug development, 6–7, 7f packaging strategies, dissolution performance
stability OOS investigation, 10 (see Dissolution performance, packaging strategies)
temperature and moisture excursions, evaluation of, 9 physical property characterization techniques, 182t
Arrhenius stability modeling, 253–254 atomic force microscopy, 184
ASAP (see Accelerated stability assessment program DSC, 183
(ASAP)) dynamic vapor sorption, 184
chemical stability, 34 isothermal microcalorimetry, 183
clinical development, 232–233 NIR and Raman spectroscopy, 184
commercial packaging and potential future changes, PXRD, 183–184
238–239, 238t SSNMR spectroscopy, 185
conversion processes (see Degradation processes) SWAXS, 185
CTAs, 232 physical stability processes, 34
drug substance synthetic route, change in, 235, 236t postapproval change submissions, 236–237
initial use period assignment, ASAP data, 233–235 potential opportunities, 239–240
stability-related CTA queries, 236, 236t practical considerations (see Practical considerations,
data processing APS study)
humidity, irregular effects of, 196–198 prediction accuracy and precision, 204, 205f
non-Arrhenius effects of temperature, 195–196 principles of, 4–5
degradation rate (see Degradation rate, APS study) product understanding and modeling algorithms, 232
design of experiment approach, 34, 232 retest period, 231–232
drug product composition selection, 239, 239t shelf-life prediction, basic data processing, 63–64, 63t
drug product use period assignments, 232 SLLA, 232
drug substance containing different particle sizes, solid forms changes
237–238 cocrystal breakback/formation, 179
effects of moisture, Arrhenius model, 148–149 deliquescence, 180, 181t
embedding new process within business (see Embedding APS evaporation and sublimation, 181–182
within business) hydration/dehydration, 178
enabled robust stability protocol design, 232 melting, 181, 181t

471
472 Index

Accelerated predictive stability (APS) (Continued) at desired temperature inside desiccator with saturated salt
polymorph conversion, 178 solutions, 265–267, 266f, 268–269f
process-induced disorder/amorphous content, 179–180 at desired temperature in temperature- and humidity-
salt disproportionation/salt formation, 178 controlled oven, 265
solvate drug product, desolvation of (see Desolvation, DMSO) humidity-corrected Arrhenius equation, 259–260
stability durations and timepoints, selection of, 57–58 pre-equilibration phase, 264, 264f
stability testing, purpose of, 231–232 relative percent errors, 260–261t, 261–262
statistical methods, 64–67 sample at room temperature in foil bags, 267–270, 270t
storage conditions, 232 temperature error, 259–260, 260t
temperature and humidity conditions, selection of, 56–57 thermal and humidity equilibration, 259–260, 264
thermal stress, 148 shelf life (see Initial shelf life prediction)
vs. traditional long-term stability studies, 176 study design, 254–256, 255t
workflow, 76, 77f unstable drug candidate, early development (see Unstable
Accelerated stability assessment program (ASAP), 85–86, 233, drug candidate, ASAP application)
253–254 water activity
ASAPprime® software, 4–6, 254–255, 271 Clausius-Clapeyron equation, 262–263, 263f
chemical stability screening and prediction, 253–254 equilibrium relative humidity, 262
drug products measurements, 262
lyophilized drug product, 276–279 working group, 271
solid oral drug product, 279–281 Accelerated stability modeling (ASM), 4, 411–412, 449
drug substance, starting materials, and intermediates drug product disintegration time, sodium bicarbonate
compound A, 272–276, 274–275t (see Sodium bicarbonate, drug product disintegration
Impurity X formation, 275, 275t times)
initial ASAP model prediction vs. long-term stability data, ionic liquids drug product (see Ionic liquids)
274, 274f nicotine lozenges, assay loss
initial stability assessments, 272 accelerated stability study, experimental design for, 450
polyethylene packaging, 272–273 analysis of, 450–456
second ASAP model prediction vs. long-term stability data, ASAPprime®, 449, 457–460
275, 275f humidity models, 450
stress-screening studies, 272, 273t kinetic models, 449–450
unmilled and milled compound A, 276, 276t long-term storage data, 457
general process flow, 259, 260t nonlinear regression, 449–450
generic drug development (see Generic drugs) NRT, 449
isoconversional model-free approach, 4 solvate drug product, desolvation of (see Desolvation, DMSO)
methodology, 270–271 Accelerated tests (ATs), 147–148
moisture modified Arrhenius equation, 4 Accelerating kinetic model, 416, 429
nicotine lozenges (see Nicotine lozenges) Active pharmaceutical ingredient (API), 306, 308, 342–345, 388
package modeling AFM. See Atomic force microscopy (AFM)
Arrhenius coefficients, 259 “AH decelerating kinetic model”, 422, 437
dynamic moisture uptake profile, 256, 257f Akaike’s information criterion (AIC), 44, 52
dynamic packaged product environment, 258, 259f Aluminum-aluminum foil blister packaging, 292
experimentally determined inputs, 258 AMBERLITE IRP64, 354, 354f
internal Lilly moisture permeability modeling tool, 256 Amlodipine (AMD) besylate tablet, 296–300
overall percent impurity prediction, 256, 258f Angiotensin-converting enzyme inhibitors (ACE), 289
predicted percent impurity change, 256, 258f Antoine equation, 195
predicted rate of degradation change, 256, 257f API. See Active pharmaceutical ingredient (API)
water activity, 256, 257f APS. See Accelerated predictive stability (APS)
postapproval environment (see Postapproval environment, AquaLab water activity meter series 3, 292
ASAP) Area percent-based method, 95
process improvements, 271 Arrhenius equation, 36–38, 36f, 253–254, 287–288, 306,
sample equilibration 323–324, 327, 330, 350, 370–371, 373t, 377, 377t,
degradation rate constant, 261–262 457–460
Index 473

Arrhenius relationship, 369–370 storage conditions, 233


Arrhenius time-scale least squares, 158, 161 unpackaged samples, 233
ASAP. See Accelerated stability assessment program stability-related CTA queries, 236, 236t
(ASAP) Cocrystals, 179
ASAPprime® software, 8–9, 65, 66f, 98, 254–255, 271, 288, Colloidal silicon dioxide (CSD), 313
291, 298, 308–311, 308t, 309f, 327, 330, 332, 334, 346, Common solid-state reaction models, 346t
347t, 358, 365, 370, 381, 386–387 Competitive degradation processes
competitive degradation processes, 187–189, 188f ASAPprime® software, 187–189, 188f
nicotine lozenges reactive environment/state, 186
assay loss, ASM, 449, 457–460 simulation, 186, 187t, 187f
modeling, 360 Consecutive degradation processes
predicted and actual shelf lives, 361–363 identical temperature and humidity sensitivities
ASM. See Accelerated stability modeling (ASM) curve shapes, 190–192
Association of South East Asian Nations (ASEAN) guidelines, simulation, 190, 190f, 191t
21, 24–25, 25t types of, 189–190
Atomic force microscopy (AFM), 182t, 184 unequal temperature and humidity sensitivities, 193–194, 193t
Atorvastatin tablets, 300–302, 301t Containers for Tablets and Capsules—Permeation method,
Autocatalytic reactions, 52–54, 53f 292–293
Avarami-Erofeyev reaction model, 346 Controlled-release matrix tablet, 339–340
Avrami-Erofeev transform, 450–454, 454t, 456f Cooks distance, 57
Critical material attributes (CMAs), 384
B Critical process parameters (CPPs), 384
Critical quality attribute (CQA), 384
Bayesian information criteria (BIC), 44, 52, 406, 416, 422,
Critical relative humidity (CRH), 79–80, 79–80t, 180, 181t, 342,
429, 450
385
Benazepril hydrochloride tablets, 384–385
Critical-to-quality (CtQ), 147–148
Blister packaging, 339–340
Crosspolarization (CP), 185
Brunauer-Emmet-Teller (BET) equation, 122
CTAs. See Clinical trial applications (CTAs)

C
Charged aerosol detection (CAD), 96 D
Chilled-mirror dew-point technology, 292 Data analysis, shelf life estimation
Clausius-Clapeyron equation, 128–129, 262–263, 263f accelerated shelf-life model, 170–172, 170t, 171f
Clinical stability studies, regulatory guidance experimental design space, 168, 169t
gaining product knowledge, 18 linear interpolation, 155–157, 156f
GMP regulations, 17 natural logarithm of shelf life, 156–158, 157f
ICH guidances, 17–18 temperature and relative humidity, 150t, 155–156
phase 1 to 3 clinical supplies, stability requirements for, APS data generation, 153–154
18–19 Arrhenius time-scale least squares, 158
stability data, 16–17 classical kinetic model
storage conditions and time points, 19–20 unweighted second-stage approach, 159
Clinical trial applications (CTAs), 232 weighted second-stage approach, 160
drug substance synthetic route, change in, 235, 236t common intercept multiple slope regression model
initial use period assignment, ASAP data unweighted second-stage approach, 160
“ICH-like” stability programs, 233 weighted second-stage approach, 161
immediate-release solid oral dosage tablet formulations, data censoring, 163
233 data rounding, 163
initial 12-month clinical use period/retest period without extended nonlinear regression model, 161–162, 167, 168t,
queries, 233–235 170–172, 170t
regulatory submissions, 233, 234t model-fitting methods, results of, 163–166
RRT prediction, 233–235, 234–235f truncated 44 factorial design (Insert symbol), 162, 162t
shelf life-limiting degradants, 233–235, 234t two-stage approach, 155
474 Index

“Decelerating kinetic model”, 416–417 packaged product, RH prediction, 388


Degradation processes, 149 product sorption effect on RH, 386–388
competitive processes IR tablets, high-humidity sensitivity
ASAPprime® software, 187–189, 188f commercial formulation development, 388–390
reactive environment/state, 186 HDPE bottles, 388–390
simulation, 186, 187t, 187f humidity modeling, 391
complex processes, 194 manufacturing of tablets, 390–391
consecutive processes (see Consecutive degradation testing, 391
processes) packaging design space, 396–399
reversible processes, 189 softest tablets, 392
solid-state reactions, 149–152, 150t stressing tablets, 392
Degradation rate, APS study temperature and humidity, drug release, 384–385
combined temperature and humidity model Dissolution prediction
humidity raised to power of constant, 41–42 accelerated experimental conditions, 375
model selection, 42–44 ASAPprime® software, 374–375
relative humidity, 40–41 data analysis
external/internal factors, 35 ASAP data inputs, 377
humidity, 38–39 dissolution modeling hypothesis, 376–377
linear degradation model, 45–46 modeling results, 377
nonlinear degradation profiles passing specification, predicted probability of, 378
complex chemical degradation mechanisms, 46 methods, 375
degradation profile shapes, 46–48, 47f model verification
heterogeneous physical states, 46 predicted %dissolution vs. real-time stability data, 381
isoconversion methods (see Isoconversion approach) tablet water content, packaged product on stability, 380
kinetic model approach, 48–50 product’s bioavailability, 374–375
limit of degradation, 46 under stressed conditions, 376, 376f
nonfixed humidity environment, 46 tablet water content, 374–375
nonlinear physical and solid-state processes, 46 Drug formulation development
shape parameter approach, 50–54, 51t, 53f controlled-release matrix tablet, 339–340
oxygen level, 44–45, 45f data analysis and modeling, 327
physical state effects, 177–182, 177f degradation rate, 323–324
temperature, 36–38, 36f experimental setup
Design of experiment (DOE) approach, 34, 232 ASAP samples, 325–327
Desolvation, DMSO temperature/humidity control and recording,
API, 463–464 stress stations with, 324–325
content analytical method, 463 in-house stress stations, 328
drug product “M” ASM experiment liquid formulations, 331–333
experimental data, 464, 465f lyophilized formulation
kinetic model fitting and selection process, 465–466, ASAP study, 335–338
465–466f drug substance ASAP study, 333
long-term stability (model) and actual stability (data), prelyophilization solution ASAP study, 334–335
466–468, 467–468f processing stages, 333
temperature-humidity experimental design, 464, 464f shelf-life prediction, 324
Differential scanning calorimetry (DSC), 182t, 183 single accelerated stability studies, 323–324
Diffusion reaction model, 346 suspension formulation, 328–330
Dimethylsulfoxide (DMSO), desolvation. See Desolvation, DMSO Drug product development
Dissolution performance, packaging strategies drug product performance, 210
CQA, 384 early drug product development (see Early clinical drug
hardest tablets, 392 product development, APS testing)
humidity changes in excipient compatibility study, 209–210
desiccants effect on RH, 388 manufacturability, 210
moisture protection, 386 Dry granulation process, 390–391
Index 475

direct compression approach, 212 Equilibrium relative humidity (ERH), packaging simulations,
excipients, 239, 239t 106
platform formulation chemical degradation rates, 106
compositions, 213, 213t degradation curve shape, 138–140
publications, 212–213 instantaneous humidity, 136–140, 137t
selection process, 214, 215f function of time, 106–107, 107f
roller compaction simulation, 212 humidity excursions, stability risks of, 144
Dynamic moisture sorption microbalance (DMSM), 291 LDPE bags, 115, 116f
Dynamic vapor sorption (DVS) technique, 110–111, 111f, 120, shelf life–limiting attributes, effects on, 144–145
128, 182t, 184, 356–357, 377, 386–387 starting value, estimation of, 109
water activity measurements, 109–110
water content, moisture sorption isotherms
E BET equation, 122
Early clinical drug product development, APS testing DVS technique, 110, 120
advantages, 208 entire packaging contents, 124–127
challenges, 208–209 GAB equation, excipients and desiccants, 120–124, 121f,
chemical incompatibilities, 210 123–124t
FAST formulation approach (see Focused accelerated headspace (air), 119–120
scientific testing (FAST) approach) hygroscopic amorphous drug substance, KF analysis,
goals in, 207–208 111–112, 111f
Electrospray ionization (ESI) interface, 327 Langmuir isotherm, desiccants, 120–122, 121f
Embedding APS within business sorption, desorption, and hysteresis, 127–128
associated gaps/risks, 244–245 temperature, effect of, 128–129
business process construction water activity of product, calculation of, 129–130
appropriate action, 252 Excel kinetic analysis, 347t
design of experiments, 247 Excipient compatibility study, 209–210
models/outcomes interrogation, 252 Expanded Arrhenius model, 153
stability models, 252 “Exp RH Decelerating” model, 450
stability protocol execution, 251 Extended Arrhenius model, 159–160
standardization, 251 Eyring equation, 38
workflow, 251
current stability practices, 244–245
development process, 244–245 F
implementation roll-out approach First-in-human (FIH) clinical trials, 312
“The Chasm”, 248–250 First-order kinetic model, 50
“The Gap”, 248–250 First-order reaction model, 346
lead user model, 248–250, 249f Fisher Scientific Isotemp incubators, 324–325
software package, 250 Fixed-dose combination (FDC) tablet, 374–375
perceived risk, 245–246 Fluid bed-drying process, 345–349
reframing, 246 Focused accelerated scientific testing (FAST) approach
risk aversion, culture change, 243–244 APS stability testing
risk mitigation strategy, 246 advantages, 217–218
stakeholder efficiency gains, 216, 216f
budget holders, 247 four- (or five-) point APS study, 216–217, 217t
internal regulatory, 248 goal, 214–215
manufacturing organization, 248 roller compaction performance, 215–216
map, 246, 247f BCS Class I compound
packaging groups, 248 APS stability data, 221, 221t
product development departments, 247–248 granule particle size distributions, 219, 220f
quality assurance group, 248 hardness vs. compression profile, 219, 220f
senior sponsors, 246 long-term stability data at ICH conditions, 221, 221t
statisticians/internal modeling experts, 247 SF-TS relationship, 219, 219f
476 Index

Focused accelerated scientific testing (FAST) approach H


(Continued) Handerson-Hasselback equation, 354, 354f
BCS Class II compound, amorphous polymer-based Hard gelatin capsule (HGC), 312
dispersion Headspace moisture analyzer model FMS-1400H, 327
granule particle size distributions, 224–226, 225f Heat induction seals (HIS), 116–118, 134
hardness vs. compression profile, 226, 226f High-resolution mass spectrometry (HRMS), 339
ICH chemical stability, 227, 227–228t Hydroxypropyl beta cyclodextrin (HPBCD), 315
physical stability, 226, 227t Hydroxypropyl methylcellulose (HPMC), 312, 339
SF-TS relationship, 224, 225f
temperature/humidity conditions, 226, 227t
BCS Class I/II compound, moderate drug loading I
APS stability data, 222, 223t Initial shelf life prediction
granule particle size distributions, 222, 223f control strategy, 381–382
SF-TS relationship, 222, 222f degradation product analysis, 371
stability data at ICH conditions, 223, 224t early development stage, 370
characterization tests, 211 experimental conditions, 371
fit-for-purpose paradigm, 211 investigational drugs, tablet formulation, 370
nonstandard excipients, 218 isoconversional approach, 370
platform formulations packaging configurations, 371–374, 374t
dry granulation (see Dry granulation process) International Conference on Harmonization (ICH) guidance
intragranular vs. extragranular lubrication, 214 clinical stability studies, 16–18
publications, 212–213 registration stability, 20–23, 22t
precautions, 218–219 Ionic liquids
standardized formulation platforms, 211–212 chromatographic results, 416
Foil-foil blisters, 293, 299–300, 386 data analysis and modeling, 416–445
Food and Drug Administration (FDA), 300 glutaric acid formulation
FreeThink Technology, 360 accelerated stability experimental results for, 414t
analysis of, 417
lactic acid formulation
G accelerated stability experimental results for, 415t
GAB equation. See Guggenheim-Anderson-de Boer (GAB) analysis of, 417–445
equation materials, 412
Gas chromatography (GC)-based methods, 96 methods
Gaussian distribution, 153, 198–199 analytical methodology, 413
GCC guidelines. See Gulf Cooperation Council (GCC) experimental methodology, 412–413
guidelines Isoconversional model-free approach, 4
Generic drugs Isoconversion approach
experimental design, 342 advantage of, 54
granulation/drying process change on stability, 345–349 degradation level of interest, 54, 54f
milling process, API, 342–345 disadvantage of, 54
preservative system degradation route, liquid formulation, interpolation and extrapolation, 55
349–351 isoconversion time, 54–55, 54f
Geometric contraction model, 149–150, 150t time to failure, 54
Glass transition, 80–82, 81f zero-order kinetics, 55
Glass transition temperature, 179–180 Isoconversion principles, 187–189, 191
Glutaric acid formulation, 412 Isothermal calorimetry, 209
accelerated stability experimental results for, 414t Isothermal microcalorimetry, 182t, 183
analysis of, 417
Good manufacturing practices (GMP), 17
Guggenheim-Anderson-de Boer (GAB) equation, 120–124, K
121f, 123–124t, 126–127, 291, 298, 356–357, 377, Karl Fischer (KF) analysis, 111–112, 111f
386–387 Kinetic model
Gulf Cooperation Council (GCC) guidelines, 21, 26 drug product “M” ASM experiment
Index 477

actual vs. predicted, 466, 466f crystalline solids, hydration and dehydration of, 82, 83f
parameter estimates, 466, 466f deliquescent excipients, CRH of, 79–80, 79–80t
selection process, 465–466, 465f melting point, 78, 78t
nonlinear degradation profiles, 48–50 physical changes, 76–77
temperature-/humidity-induced polymorph conversion, 83
Maximum daily dosage (MDD), 350–351
L Mean kinetic temperature (MKT), 9, 36
Lactic acid formulation, 412 Microcrystalline cellulose (MCC), 339
accelerated stability experimental results for, 415t Minitab software package (version 16), 302
analysis of Moisture modified Arrhenius equation, 4
assay data, 417–422 Moisture permeability, packaging simulations
“Deg9” degradation product, 437 MVTR/WVTR, 107, 112–113
N-oxide degradation product, 422 permeability
RRT 0.51 degradation product, 422 applications, 113–114
RRT 0.72 degradation product, 422–428 bottle size, 118
RRT 0.84 degradation product, 429 definition, 113
RRT 0.90 degradation product, 429 example coefficients, 114, 114t
RRT 0.97 degradation product, 429–436 HIS for bottles, 116–118
RRT 1.22 degradation product, 437 intrinsic permeability, 113–114
RRT 1.35 degradation product, 437 LDPE bags, 114–115
RRT 2.02 degradation product, 437–445 multiple packaging layers, 115–116
storage conditions, optimization of, 446–448 Moisture sorption isotherms, 291–292, 356–357
Langmuir isotherm, 120–122, 121f ERH and water content, packaging simulations
Lean stability strategy, 16 BET equation, 122
clinical development, 30 DVS technique, 110, 120
global reception, 31 entire packaging contents, 124–127
line extensions and postapproval changes, 31 GAB equation, excipients and desiccants, 120–124, 121f,
registration, 30–31 123–124t
science and risk based, 30 headspace (air), 119–120
SRQA and SLLA, 30 hygroscopic amorphous drug substance, KF analysis,
Limit of detection (LoD), 151 111–112, 111f
Limit of linearity (LoL), 151 Langmuir isotherm, desiccants, 120–122, 121f
Limit of quantitation (LoQ), 151 sorption, desorption, and hysteresis, 127–128
Linear Arrhenius model, 9 temperature, effect of, 128–129
Linear degradation model, 45–46 water activity of product, calculation of, 129–130
“Linear Time Kinetic Model”, 416, 422 nicotine lozenges, 356–357
Liquid formulations, 331–333, 332t pharmaceutical solids, 297–298
LTQ Orbitrap, 327 Moisture vapor transmission rate (MVTR), 8–9, 100, 107,
Lyophilized drug product, 276–279 112–113, 238, 238t, 292–293, 384–386
Lyophilized formulation, 276–278, 278t Monte Carlo method, 157–158, 198–199, 293, 298, 302
ASAP study, 335–338 Multilinear regression analysis, 40–41, 52
drug substance ASAP study, 333 Multinonlinear regression analysis, 40–41, 53–54
prelyophilization solution ASAP study, 334–335
processing stages, 333
N
Near-infrared spectroscopy (NIR), 182t, 184, 190
M Nicotine lozenges
MadgeTech, 356 ASAPprime® modeling, 360
Marketing authorization (MA), 236–238 ASAP study design, 356
Mass spectrometry detection (MSD), 96 assay loss, ASM
Material attributes, APS study accelerated stability study, experimental design for, 450
amorphous drugs, glass transition and crystallization, 80–82, analysis of, 450–456
81f ASAPprime®, 449, 457–460
478 Index

Nicotine lozenges (Continued) intrinsic permeability, 113–114


humidity models, 450 LDPE bags, 114–115
kinetic models, 449–450 multiple packaging layers, 115–116
long-term storage data, 457 steps for, 130–131
nonlinear regression, 449–450 typical process for, 108–109, 108f
NRT, 449 user-intervention events
bioavailability of, 354 adding/refreshing desiccant, 134
desiccant strip (molecular sieve), 354–355 air exchange, 133–134
dosage forms, 353 HIS bottle, permeability of, 134
HPLC, 358 removing product/desiccant, 134
instability, 366 Parahydroxybenzoic acid (PhBA), 350
isoconversion points estimation, 358–359 Permeability, packaging simulations
lipophilic mucosal tissues, 354 applications, 113–114
long-term stability/nicotine assay of, 355f bottle size, 118
moisture sorption isotherm, 356–357 definition, 113
oromucosal tissues, 354 example coefficients, 114, 114t
pH, 353 HIS for bottles, 116–118
polymethyacrylate polymer, 354f intrinsic permeability, 113–114
polypropylene vial, 355f LDPE bags, 114–115
predicted and actual shelf lives multiple packaging layers, 115–116
calculation, 361–363 Pharmacokinetics (PK), 384
discrepancies, 363–365 Photodiode array detection (PDA), 96
PVdC blisters, 354–355, 355f PIB. See Powder-in-bottle (PIB)
stability limit, 358 PIC. See Powder-in-capsule (PIC)
temperature and humidity, 355–356 Polychlorotrifluoroethylene (PCTFE), 386
Nicotine polyacrilex (NPA), 354, 354f Polymer excipients, 81–82
Nicotine replacement therapy (NRT), 353 Polynomial equations, 291
NIR. See Near-infrared spectroscopy (NIR) Polyvinyl chloride (PVC), 386
Nonlinear regression model, 158, 161–162 Polyvinylidine chloride (PVDC), 386
Nonmoisture-equilibrated tablets, 375 Postapproval environment, ASAP
Nucleation models, 149–150, 150t amlodipine besylate tablet, 296–300
Arrhenius kinetics, 287–288
O commercial space, 288
degradation kinetics, 288
Optical microscopy (OM), 182t, 184
ICH conditions, 289
Out-of-specification (OOS) stability, 288–296
in-processed samples, sampling scheme for, 295t
magnesium carbonate, 289, 294
P multilinear regression approach, 288
Packaging simulations OOS stability issues, 289–296
accuracy assessment, experiment, 135–136 packaging configuration changes, 300–302
bottle count, moisture protection, 141, 142f pharmaceutical production, 288
equilibrium relative humidity (see Equilibrium relative proposed package change, 299–300, 302
humidity (ERH), packaging simulations) stability data, 288
in-use stability study, 142–143, 143f temperature and relative humidity, 287–288
multiple packaging layers, 132–133 water activity, 292
MVTR/WVTR, 107, 112–113 Postapproval Stability Protocol and Stability Commitment,
packaging selection, 141 236–237, 239
permeability Powder for oral solution (PfOS) prototype formulations
applications, 113–114 capsule shell, PIC assessment, 312
bottle size, 118 excipients effects and capsule shell, 313–314
definition, 113 Powder-in-bottle (PIB), 312, 314t
example coefficients, 114, 114t Powder-in-capsule (PIC), 312, 314t
HIS for bottles, 116–118 Powder X-ray diffraction (PXRD), 182t, 183–184
Index 479

Power model, 416 Reaction order model, 149–150, 150t


Practical considerations, APS study, 76, 77f Registration stability studies
analytical testing methods, 96t aim of, 20
appearance/description, 95–96, 96t ASEAN guidelines, 21, 24–25, 25t
artificial degradation product, 97 data analysis, 28
assay and degradation products, 96, 96t GCC guidelines, 21, 26
degradation product, identification of, 96–97, 96t ICH guidance, 20–23, 22t
physical tests, 96, 96t lean stability strategy, 30–31
report of results, 97 minimum data requirements, principles for,
suitability of, 95 27–28
data fitting, 97–100, 99–100t postapproval stability requirements
material attributes, evaluation of changes, classification of, 29, 29t
amorphous drugs, glass transition and crystallization, commitment, 28
80–82, 81f regional/country-specific guidance documents, 21,
crystalline solids, hydration and dehydration of, 82, 83f 26–27
deliquescent excipients, CRH of, 79–80, 79–80t stability testing, purpose of, 20
melting point, 78, 78t WHO guidelines, 21, 23–24, 24t
physical changes, 76–77 Regulatory stability guidance
temperature-/humidity-induced polymorph clinical stability studies
conversion, 83 gaining product knowledge, 18
moisture sorption/desorption isotherm of product, 100 GMP regulations, 17
MVTR of packaging, 100 ICH guidances, 17–18
protocol design phase 1 to 3 clinical supplies, stability requirements
comprehensive approach, 90, 90t for, 18–19
information, 84 stability data, 16–17
isoconversion limit, 84–85 storage conditions and time points, 19–20
lean approach, 86–87, 87t lean stability strategy, 16
storage conditions, time points, and repeats, 85–90, 86f clinical development, 30
temperature and relative humidity limits, 84 global reception, 31
two-step approach, 87–90, 88t, 89f line extensions and postapproval changes, 31
sample storage, 90–91 registration, 30–31
and pull schedule, 94–95 science and risk based, 30
saturated salt solution, use of, 91, 92t, 93f SRQA and SLLA, 30
temperature and RH stability chambers, use of, 91–94, registration (see Registration stability studies)
93t, 94f Relative humidity (RH), 106, 292
Prediction error sum of squares (PRESS), 67 degradation rate, 37–42
Prior approval supplement (PAS), 237 limits, 84
Process-induced disorder, 179–180 packaged products
Prout-Tompkins data transformation, 450–454, 454t, 455f, ERH (see Equilibrium relative humidity (ERH),
457–460 packaging simulations)
Prout-Tompkins model, 149, 150t moisture sorption/desorption isotherm, 100
Pseudo-zero-order model, 159 MVTR, 100
PXRD. See Powder X-ray diffraction (PXRD) stability chambers, use of, 91–94, 93t, 94f
PyroButton-SQL software, 325, 328 Relative standard deviation (RSD), 199–200
Resodyn Acoustic Mixer (RAM), 316–317
Q Reversible degradation processes, 189
RH accelerating model, 453
Quality-by-design (QbD) perspective, 384–385
RH linear time model, 450
Quinapril tablets, 289–292, 291t, 294–296
“RH Power” model, 422, 453
Risk-based predictive stability (RBPS), 5–6
R Risk mitigation strategy, 246
Raman spectroscopy, 182t, 184 Root mean square error (RMSE), 66
Raoult’s law, 180 Ross equation, 80
480 Index

S trona hypothesis, 409


Sample storage, APS study, 90–91 Sodium dodecyl sulfate (SDS), 326
and pull schedule, 94–95 Solid fraction tensile strength (SF-TS) relationship
saturated salt solution, use of, 91, 92t, 93f BCS Class I compound, 219, 219f
temperature and RH stability chambers, use of, 91–94, 93t, BCS Class II compound, amorphous polymer-based
94f dispersion, 224, 225f
SAXS. See Small-angle X-ray scattering (SAXS) BCS Class I/II compound, moderate drug loading, 222,
Scanning electron microscopy (SEM), 404 222f
Second-order reaction model, 346 Solid oral drug product, 279–281
SF-TS relationship. See Solid fraction tensile strength (SF-TS) Solid oral formulation (SOF) assessment, 311–312,
relationship 314–315
Shape parameter approach, 192 Solid-state NMR (SSNMR) spectroscopy, 182t, 185
AIC and BIC, 52 Sorption-desorption moisture transfer model (SDMT), 291
Avrami-Erofeyev process, 50–51 SOTAX DT 2 Disintegration Tablet Tester, 391
curve shapes, 50, 51f Stability-related material attributes (SRMAs), 7–8, 11–12
limitation, 51 Stability-related quality attributes (SRQA), 30, 237
multilinear regression, 52 Suspension formulation, 328–330
sigmoidal and exponential kinetics, 52–54, 53f SWAXS. See Small- and wide-angle X-ray scattering
solid-state degradation processes, kinetic expressions for, (SWAXS)
50–51, 51t
Shelf life T
data analysis (see Data analysis, shelf life estimation) Tablets development
definition, 147–148 API-excipient binary system, 314
initial shelf life (see Initial shelf life prediction) binary excipient compatibility study, results of,
predictions 315–316t
atorvastatin tablets, 300–302 degradation-free excipients, 314–315
based on 30ºC/75%RH conditions, 293f, 295f prototype tablet formulations
confidence intervals, 293, 302 basic stabilizers, 320–321
degradant A, 296, 298 common excipients, 317–319
degradant B, 302 SOFs, 314–315
extrapolations, 293, 299, 302 tablet stability process, 316–317
packaging, 299–300 Thermogravimetric analysis (TGA), 183
quinapril tablets, 289–290, 292, 295–296 Trifluoroacetic acid (TFA), 326
real-time data, 289 Turbula-blending process, 316–317, 317t
stability data, 288
Shelf life-limiting attributes (SLLAs), 5, 7, 30, 232
Sigmoidal degradation profiles, linearization of, 450–456
U
Unstable drug candidate, ASAP application
Simulations, packaged products. See Packaging simulations
Arrhenius equation, 306
Single-pot process, 345–349
drug product stability assessment
Small- and wide-angle X-ray scattering (SWAXS), 182t, 185
PfOS prototype formulations, 312–314
Small-angle X-ray scattering (SAXS), 182t, 185
tablets development, 314–321
Sodium bicarbonate, drug product disintegration times
drug substance stability assessment
accelerated stability data, 405
API initial retest period, 307–309
activation energy, 404
ICH stability data, 309–310
analysis of, 405–407
long-term storage condition, 306–307
curvature types, 404
model-free isoconversional kinetic approach, 306
decomposition reaction, 403–404
short single temperature stress testing protocol, 306
humidity mechanisms, 404
stability testing, 305
long-term disintegration time, 408–409
primary accelerated stability study experimental
design, 405 V
SEM observation, 404 Vapor pressure (VP), 39, 41–42
Index 481

W X
Water vapor transmission rate (WVTR). See Moisture vapor XLfit software, 98, 98f
transmission rate (MVTR)
Wet granulation process, 212, 289, 294–296, 385 Z
Whole-vial-assay approach, 413 Zero-order reaction model, 346
World Health Organization (WHO) guidelines, 21, 23–24, 24t

You might also like