SUMMARY - Questions Flashcards

(292 cards)

1
Q

What are the primary objectives of cancer screening?

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

How are the validity of screening and the validity of a screening program evaluated?

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What are some common cancer screening methods mentioned, including the target age ranges and frequencies?

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Explain the role of Circulating Tumor Cells (CTC) and Circulating Tumor DNA (CTDNA) as metastatic cancer biomarkers.

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What are the clinical implications associated with detecting CTC and CTDNA?

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What is a biomarker defined as?

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What are some pros and cons of using liquid biopsy (blood sample) as an alternative to solid biopsy?

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Describe the different types of biomarkers based on their clinical application (e.g., predictive, prognostic, diagnostic) and provide an example for each.

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

List some of the molecular markers that can be detected in blood, urine, or saliva.

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Discuss the challenges associated with using blood/serum and urine for biomarker detection.

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What are some of the technologies used for the detection of biomarkers?

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Define and differentiate between Genomic, Genetics, Pharmacogenetics, and Pharmacogenomics (Pgx).

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What is Functional genomics, and how can it guide personalized medicine?

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Explain Comparative genomics.

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Distinguish between Germline and Somatic genomic variants and provide an example of how each can arise.

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

How is Genome instability and mutation linked to the Hallmarks of cancer?

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Why might different metastatic sites show different DNA variations compared to the primary tumour?

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What are the different types of genomic testing described?

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

How is monitoring a tumour’s genomic profile clinically applied at various stages?

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What might the detection of DNA variants prompt in terms of further investigation?

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Describe the characteristics of familial/inherited cancers with genetic pre-disposition compared to rare familial cancer syndrome with a distinct phenotype.

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Explain Knudson’s 2 hit hypothesis and how it applies to hereditary vs sporadic cancer cases.

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What types of screening are mentioned for individuals with a genetic predisposition to cancer?

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

How is Lynch syndrome diagnosed, and what is a preferred treatment approach based on diagnosis?

A

Answer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
How does germline data inform treatment decisions in pharmacogenomics?
Answer
26
Why is pharmacogenomics considered better than a standard approach in some cases?
Answer
27
Explain the allele category and diplotypes system used to describe enzyme metaboliser status.
Answer
28
Provide examples of how prodrugs (like codeine) and specific enzymes (like CYP2D6 and UGT1A1in Gilbert syndrome) are affected by genetic variation, and the clinical implications.
Answer
29
What inherited variants in the DPYD gene are mentioned, and why is DPYD testing important for patients receiving Fluoropyrimidines?
Answer
30
Discuss the importance of diversity in genomic variation for treatment decisions and the existing gap in genomic data.
Answer
31
Provide examples of how pharmacogenomics can personalize healthcare for specific drugs like Clopidogrel, Carbamazepine, and Statins.
Answer
32
Name some genes and the drugs for which PGx guidelines are available from CPIC.
Answer
33
What are some key challenges in the field of personalized medicine omics?
Answer
34
Define Proteomics, Clinical proteomics, and Translational proteomics.
Answer
35
What is the primary technology/tool used for proteomic detection and analysis?
Answer
36
Describe the Ionization techniques used in mass spectrometry (Electrospray and MALDI).
Answer
37
Define Metabolomics and explain how metabolites can act as a chemical "fingerprint".
Answer
38
What are the main challenges in Metabolomics?
Answer
39
Explain the difference between Untargeted and Targeted Metabolomics techniques.
Answer
40
What technologies are used for metabolomic detection and analysis?
Answer
41
Outline the strategies for drug target discovery using omics techniques.
Answer
42
How does Multi-omics provide a comprehensive view in drug discovery?
Answer
43
What is the clinical relevance of omics, particularly in understanding the need for precise, molecularly targeted therapies?
Answer
44
Differentiate between Targeted therapy and Chemotherapy.
Answer
45
How do techniques like Next generation sequencing, gene expression analysis, and the Human epigenome project (HEP) contribute to understanding cancer biology and guiding treatment?
Answer
46
How do Epigenetic Drugs that target chromatin work, and how are they used in combination therapies?
Answer
47
Compare Traditional cancer drugs with Chromatin-targeted drugs in terms of their mechanisms and toxicity.
Answer
48
How can molecular profiling and companion diagnostics help direct the use of targeted agents beyond standard of care?
Answer
49
What is the role of patient selection in ensuring effective treatment and minimizing harm with targeted therapies?
Answer
50
What are the updated hallmarks and enabling characteristics of cancer, and how do they form the basis for targeted therapies?
Answer
51
Identify the hallmarks considered REQUIRED for a cell to become cancerous vs those that are NOT REQUIRED but contribute to cancer progression.
Answer
52
How does the inflammatory microenvironment support cancer?
Answer
53
Explain how Obesity increases cancer risk based on the information provided.
Answer
54
Describe how cancer cells Deregulate cellular metabolism to meet their high energy and biosynthesis demands.
Answer
55
Explain the Warburg effect and how cancer cells adapt to its toxic byproduct.
Answer
56
How do Genetic and Epigenetic changes influence gene expression in cancer cells?
Answer
57
How do the Tissue origin and the Tumor microenvironment influence cancer cell metabolism?
Answer
58
List and briefly explain some of the key evasion strategies cancer cells use for Avoiding immune destruction.
Answer
59
How do cancer cells hijack inflammation for their benefit?
Answer
60
Explain how Genome instability and mutation arise in cancer cells and lead to tumour heterogeneity and metastasis.
Answer
61
What is Non-mutational epigenetic reprogramming, and what are its key mechanisms?
Answer
62
How does DNA Methylation (hypermethylation and hypomethylation) impact gene expression in cancer?
Answer
63
What are Histone Modifications, and how do they affect chromatin structure and gene expression?
Answer
64
In which cancer types are abnormal methylation patterns commonly found?
Answer
65
Provide examples of methylation-based biomarkers and their prognostic or diagnostic roles in specific cancers.
Answer
66
How do Polymorphic microbiomes around tumors influence cancer?
Answer
67
How do kinases, particularly receptor tyrosine kinases (RTKs), function in normal cell signaling?
Answer
68
What are some mechanisms by which dysregulated RTKs contribute to cancer?
Answer
69
Describe the intracellular kinase pathways mentioned (e.g., RAF-MEK-ERK) and their role in cancer.
Answer
70
How do EGFR inhibitors work, and what are some common resistance mechanisms to them?
Answer
71
Explain the mechanism of Vemurafenib as a BRAF inhibitor and why resistance often occurs.
Answer
72
How does radiation damage DNA through direct and indirect mechanisms?
Answer
73
Why are hypoxic tumors resistant to radiotherapy?
Answer
74
Describe the difference between External beam radiotherapy (X-ray/MRI vs Proton beam).
Answer
75
Explain the mechanisms of Internal radiotherapy (Brachytherapy and Radiopharmaceutical therapy) using examples like Radium-223 and Lutetium isotopes.
Answer
76
How does combining Radiotherapy with chemotherapy enhance the response, using Cisplatin, 5-Fluorouracil (5-FU), and Gemcitabine as examples?
Answer
77
What is the role of the MGMT enzyme in the response to Temozolomide in glioblastoma, and how is its methylation status used as a predictive biomarker?
Answer
78
Explain the mechanism of Immune checkpoint inhibitors, such as those targeting PD1/PDL1.
Answer
79
How can radiotherapy and chemotherapy potentially influence PDL1 expression?
Answer
80
What is the concept of Therapeutic ratio when combining chemo and radiotherapy?
Answer
81
What are some key challenges related to emerging resistance in highly targeted therapies?
Answer
82
Using ALK inhibitors as an example, describe the mechanism, initial effectiveness, resistance mechanisms, and strategies to overcome resistance.
Answer
83
Why do cancers with a high mutation burden often respond better to immunotherapy?
Answer
84
Explain the role of KRAS in signaling pathways and the strategies attempted to target it, including the new approach using Storasil for the KRAS G12C mutation.
Answer
85
Compare using SiRNA and CRISPR as genetic tools to block protein production like KRAS.
Answer
86
Explain the concept and mechanism of PROTACs (PROteolysis TArgeting Chimera), drawing on the ubiquitin proteosome system.
Answer
87
How can PROTACs be attached to antibodies?
Answer
88
Describe the evolution of PROTACs from the Ovacillin example to small molecule PROTACs, highlighting the key improvements and advantages.
Answer
89
Using ARV-110 and ARV-471 as examples, explain how small molecule PROTACs target specific receptors (Androgen Receptor and Estrogen Receptor).
Answer
90
Why are PROTACs considered better than traditional endocrine therapies like Tamoxifen or Fulvestrant?
Answer
91
Summarize the key Advantages and Challenges of using PROTACs.
Answer
92
What are Antibody Drug Conjugates (ADCs), and how are they designed to deliver chemotherapy more safely?
Answer
93
Describe the different regions of an antibody (FAB and FC) and their roles in ADC function.
Answer
94
Explain the mechanism of action for ADCs.
Answer
95
What are the different types of linkers used in ADCs, and what are the potential issues if the linker breaks prematurely?
Answer
96
List the different types of payloads (cytotoxic drugs) used in ADCs.
Answer
97
What is the Drug to antibody ratio (DAR)?
Answer
98
Using Kadcyla (TDM1) and ENHERTU (TDX2) as examples, explain how ADCs are structured and why one might be considered better than the other.
Answer
99
What are the main drawbacks and problems associated with ADCs, including off-target toxicityand poor tumor penetration?
Answer
100
Despite the drawbacks, what is a noted advantage of ADCs?
Answer
101
Explain the concept of Peptide drug conjugates (PDCs) using the CMML example and targeting CCR2 receptors.
Answer
102
Why might chemotherapy not be suitable for CMML cells based on the source?
Answer
103
What are the potential benefits of PDCs?
Answer
104
Compare the potential advantages and disadvantages of Antibody Drug Conjugates, Antibody Degrader Conjugates, and Peptide Drug Conjugates.
Answer
105
How can the concept of mRNA vaccines used for viruses be applied to cancer targeting?
Answer
106
Describe the different ways mRNA can be delivered, including the use of lipid-based nanoparticles.
Answer
107
Explain the Mechanism of Action for formulated mRNA delivered via lipid nanoparticles, and how it can reactivate the immune response against tumors.
Answer
108
What are lipid nanoparticles made of, and what is the role of each component?
Answer
109
How do lipid nanoparticles release mRNA inside the cell?
Answer
110
Summarize the Advantages and Disadvantages of mRNA vaccines.
Answer
111
What are some future considerations for mRNA cancer vaccines, particularly regarding personalization and manufacturing?
Answer
112
Define Advanced Therapy Medicinal Products (ATMPs).
Answer
113
Define Gene Therapy, Somatic Cell and Tissue Engineering.
Answer
114
Explain the different types of viral vectors used for gene therapy (Adenovirus, AAV, Retrovirus, Lentivirus) and their characteristics (in vivo vs ex vivo, integration, immunogenicity, size capacity).
Answer
115
Outline the process of creating and using CAR T cells.
Answer
116
Provide examples of approved CAR T cell products and the types of cancers they are used for.
Answer
117
Describe the specific process for Kymriah, including how the cells are obtained, modified, and stored.
Answer
118
What are some examples of Somatic cell therapy and Tissue Engineering products?
Answer
119
Describe the common toxicities of CAR-T therapy, including Cytokine release syndrome and Immune effector cells associated neurotoxicity (ICANS), and their management.
Answer
120
What are the key implications of ATMPs for pharmacy, including governance, regulation, handling, storage, toxicity management, and patient monitoring?
Answer
121
What is lymphodepletion chemo and bridging therapy in the context of CAR-T administration?
Answer
122
Name some monoclonal antibodies (Mabs) used in Acute Leukaemia and the targets they bind to.
Answer
123
How do these Mabs kill cancer cells compared to chemotherapy?
Answer
124
What are Targeted Therapies (TKIs), and when are they used in haematological malignancies?
Answer
125
Provide an example of an Antibody-Drug Conjugate used in AML.
Answer
126
Explain how Bispecific Mabs (BiTEs) work, using Blinatumomab as an example.
Answer
127
Why might some cancers, like lung cancer and melanoma, respond better to immunotherapy?
Answer
128
List common symptoms of acute leukaemia.
Answer
129
What are some risk factors for acute leukaemia?
Answer
130
How is acute leukaemia typically diagnosed?
Answer
131
Describe the main types of Acute Leukaemia (AML, ALL, APL).
Answer
132
Outline the treatment approaches for AML, including intensive/non-intensive chemo, advanced therapies, and molecular targets.
Answer
133
Describe Acute Promyelocytic Leukaemia (APL), its cause, treatment, and prognosis.
Answer
134
What are the key characteristics of ALL, including commonality, types, and the need for CNS prophylaxis?
Answer
135
List treatment options for ALL, including chemotherapy, Mabs, TKIs, and approaches for relapsed disease.
Answer
136
Describe the causes, features, and diagnostic markers of Chronic Myeloid Leukaemia (CML).
Answer
137
How do TKIs work in treating CML, and why were second-generation TKIs developed?
Answer
138
What are some side effects and management considerations for TKIs in CML?
Answer
139
Describe Chronic Lymphocytic Leukaemia (CLL), including its cause and how gene testing informs prognosis and treatment.
Answer
140
Compare Chemoimmunotherapy and Targeted therapies for CLL.
Answer
141
Explain the mechanisms of Small molecule inhibitors used in CLL, such as BTKis (Ibrutinib, Acalabrutinib, Zanubrutinib) and BCL2is (Venetoclax).
Answer
142
What are the main side effects and management considerations for BTKis and BCL2is, particularly regarding cardiovascular risk and tumour lysis syndrome?
Answer
143
Compare the pros and cons of Continuous therapy with BTK inhibitor monotherapy vs Fixed duration venetoclax-based treatment for CLL.
Answer
144
Describe the general characteristics and diagnosis of Lymphoma.
Answer
145
Outline the treatment approach for DLBCL using R-CHOP, including the components, common side effects, and supportive drugs.
Answer
146
Explain the mechanism of Rituximab in treating Lymphoma.
Answer
147
What is used for relapsed/refractory DLBCL?
Answer
148
What is the first-line treatment for Hodgkin lymphoma?
Answer
149
Name and describe the mechanism of some drugs used for relapsed/refractory Hodgkin lymphoma.
Answer
150
What is Multiple myeloma, and what are its common clinical features?
Answer
151
How is multiple myeloma diagnosed, including the role of immunoglobulins, free light chain test, paraproteinaemia, electrophoresis, and immunofixation test?
Answer
152
Distinguish between MGUS and Symptomatic/active myeloma.
Answer
153
What are the key criteria for diagnosing active myeloma, including bone marrow findings and the CRAB features?
Answer
154
List the main treatment approaches for multiple myeloma, including transplant, immunomodulatory drugs, Mabs, proteosome inhibitors, ADCs, BiTEs, and conventional chemotherapy.
Answer
155
Explain the mechanisms of some specific treatments like Thalidomide, anti-CD38 Mabs, Proteosome inhibitors, Bilantumab mafadotin (ADC), and BiTEs.
Answer
156
What factors influence drug choice in the setting of relapsed myeloma?
Answer
157
When is Stem Cell Transplantation used, and what is its goal?
Answer
158
Describe the different types of Stem Cell Transplants (Autologous vs Allogeneic) and when each is typically used.
Answer
159
Explain the process for Autologous Stem Cell Transplant, highlighting the goal of rescuing bone marrow after high-dose chemotherapy.
Answer
160
Outline the process for Allogeneic Stem Cell Transplant, including conditioning and ongoing immunosuppression.
Answer
161
What is the Graft-versus-malignancy (GvM) effect in allogeneic transplant?
Answer
162
What are some key considerations for donor matching in allogeneic transplant?
Answer
163
Describe the major complications of stem cell transplant, such as Graft Failure, Graft-versus-Host Disease (GvHD), Veno-Occlusive Disease, and Infections.
Answer
164
How is GvHD typically managed, and what is the challenge in balancing immunosuppression?
Answer
165
What factors are assessed for Patient Selection for stem cell transplant?
Answer
166
How do viruses infect host cells?
Answer
167
Explain the key molecular mechanisms through which viral infections can lead to cancer, including Viral Oncoproteins, Signalling Mimicry, Disruption of DDR, Chronic Inflammation, DNA Integration, and Epigenetic Changes.
Answer
168
What are some co-factors that contribute to cancer development alongside viral infections?
Answer
169
Explain how EBV causes Burkitt lymphoma, mentioning the role of viral proteins (EBNA2, EBNA3C, LMP1, LMP2, EBNA1, HBZ) and CMYC translocation.
Answer
170
How does HPV lead to cervical cancer, specifically mentioning the role of E6 and E7 proteins.
Answer
171
Explain how Hepatitis B virus (HBV) infection leads to HCC, including the role of the HBx protein.
Answer
172
How does Hepatitis C (HCV) lead to HCC, mentioning the role of viral proteins, ROS, and cancer stemness.
Answer
173
Explain how Human T lymphotropic virus-1 (HTLV-1) induces Adult T Cell Leukemia/Lymphoma, highlighting the roles of TAX and HBZ proteins.
Answer
174
What are the molecular mechanisms through which bacterial infections can induce carcinogenesis?
Answer
175
How do bacteria evade the immune system (evade complement, avoid phagocytosis, bypass antigen presentation)?
Answer
176
Explain the concept of using anaerobic bacteria for precise targeting to a tumour for delivering anti-cancer agents.
Answer
177
How do parasite infections lead to human malignancies, including their mechanisms of infecting and evading the immune system?
Answer
178
Provide examples of parasites linked to specific cancers and the mechanisms involved (ROS, inflammation, DNA damage, oncogene activation).
Answer
179
How does the immune system fight viruses?
Answer
180
Explain how vaccines, specifically the HPV vaccine, are developed based on viral mechanisms.
Answer
181
What is Oncolytic virotherapy, how does it work, and how does it relate to turning cold tumours into hot tumours?
Answer
182
How can targeting CD47 on cancer cells enhance the immune response?
Answer
183
Define PK (Pharmacokinetics) and PD (Pharmacodynamics), and explain their relationship (PK/PD).
Answer
184
Why does the relationship between concentration and effect matter in cancer therapy, especially for drugs with narrow therapeutic windows?
Answer
185
Explain the concept of variability in drug response, including predictable (intrinsic) and unpredictable (unexplained) variability.
Answer
186
Using 5-Fluorouracil (5-FU) as an example, discuss its narrow therapeutic window, variable PK, concentration-dependent toxicity, metabolism by the DPD enzyme, and the importance of DPYD polymorphism and genetic testing for dosing.
Answer
187
Differentiate between Stratified dosing and Individualised/personalised precision dosing.
Answer
188
How is Gentamicin dosing stratified for specific patient populations?
Answer
189
Explain the biotransformation of 6-MP and the role of the TPMT enzyme.
Answer
190
Describe the link between TPMT enzyme activity (including polymorphism and genotype/phenotype testing) and the efficacy and toxicity of thiopurine drugs.
Answer
191
How do Virtual Twins & Precision Dosing approaches work, and what types of data are captured for individualization?
Answer
192
What tools are used in precision dosing, such as AI and QSP models?
Answer
193
List some clinical applications of these advanced precision dosing strategies.
Answer
194
Understand how cancer therapies cause toxicity and how patient factors (like genetics, age, polypharmacy) influence risk.
Answer
195
Describe the different types of Nausea and vomiting associated with chemotherapy and their underlying pathways.
Answer
196
Explain the causes of Diarrhoea and Mucositis induced by chemotherapy.
Answer
197
How does the immune response contribute to Mucositis?
Answer
198
What are the main patient factors affecting toxicities (Intrinsic and Extrinsic)?
Answer
199
Explain the mechanism of Peripheral Neuropathy caused by platinum compounds.
Answer
200
What are the patient factors that affect the risk of chemo-induced peripheral neuropathy?
Answer
201
Describe the mechanism of Cardiotoxicity induced by Doxorubicin, including the role of ROS and iron.
Answer
202
How is late-onset Doxorubicin cardiotoxicity managed or prevented?
Answer
203
What are the patient factors and co-medications that affect the risk of doxorubicin-induced cardiotoxicity?
Answer
204
Explain the mechanism of Immune checkpoint inhibitor induced colitis.
Answer
205
What are the patient factors affecting the risk of ICI-induced colitis?
Answer
206
Explain the mechanism of BRAF and MEK inhibitors and how the BRAF V600E mutation drives uncontrolled proliferation in melanoma.
Answer
207
How do BRAF + MEK inhibitors cause skin effects?
Answer
208
What are the key challenges in traditional cancer chemotherapy and drug delivery?
Answer
209
Describe the structural and functional abnormalities of tumor vasculature.
Answer
210
Explain the Enhanced Permeability and Retention (EPR) effect and its role in passive targeting with nanomedicine.
Answer
211
What are some factors that affect the EPR effect?
Answer
212
How can the EPR effect be improved or enhanced through physical and pharmacological strategies?
Answer
213
Differentiate between passive and active targeting strategies in nanomedicine delivery.
Answer
214
What are the potential pros and cons of Active vs Passive targeting?
Answer
215
How are nanomedicines typically cleared from the body based on their size?
Answer
216
Using Doxil as an example, describe how nanomedicine can be formulated (liposome encapsulation, PEGylation) to improve drug delivery and reduce side effects.
Answer
217
Explain the challenges associated with formulating Paclitaxel and how the development of Abraxane (Nab-Paclitaxel) overcame these issues.
Answer
218
Describe the mechanism of cellular uptake for Nab-Paclitaxel, specifically mentioning Caveolin-Dependent Endocytosis and organelle targeting.
Answer
219
What are the main Biological Barriers to Drug Delivery, and how do strategies like PEGylationhelp overcome the vasculature barrier?
Answer
220
Explain the components of a Targeted Drug Delivery Strategy, including ligands, linkers, and cargo.
Answer
221
Using the SGT-53 clinical trial example, explain how liposome delivery and a transferrin ligandare used for cell targeting of a large gene.
Answer
222
Describe the concept of Organelle Targeting Strategies, particularly for Lysosomes, and the methods used for Lysosomal Escape.
Answer
223
How is Nucleus Targeting achieved, given the barrier presented by the nuclear envelope?
Answer
224
Explain the strategies for Mitochondria Targeting (passive and active).
Answer
225
What are some strategies for targeting the ER & Golgi Apparatus?
Answer
226
Describe different Advanced responsiveness strategies to improve targeted delivery based on the tumour microenvironment, such as pH-Responsive, Enzyme/MMP2 responsive, and Redox responsive drug delivery.
Answer
227
Explain the mechanisms of Magnetic hyperthermia and Light photothermal nanomaterial as stimuli-responsive strategies.
Answer
228
What are some key translational barriers in developing nanomedicine?
Answer
229
Differentiate between Supportive care and Holistic care.
Answer
230
What are some increasing complexity factors that influence patient care?
Answer
231
List the different causes of Cancer Pain.
Answer
232
Describe the general Pain Treatment Approach for cancer pain, including the use of non-opioids, weak opioids, and strong opioids.
Answer
233
Define Breakthrough Cancer Pain (BTcP) and how it is typically managed.
Answer
234
What are the common analgesics mentioned for cancer pain?
Answer
235
Summarize key considerations for Opioid Safety and potential side effects.
Answer
236
What are the criteria for referring a patient to Specialist Supportive Care for pain management?
Answer
237
Explain the concept of Psychedelic therapy for depression and anxiety in cancer patients and why they might be considered better than traditional antipsychotics.
Answer
238
What are some caveats or challenges associated with conducting clinical trials for psychedelics?
Answer
239
Describe the general pharmacology and brain effects of psilocybin.
Answer
240
Define Therapeutic Drug Monitoring (TDM) and explain its purpose and importance in clinical practice.
Answer
241
What are the main indications for TDM?
Answer
242
Explain the relationship between the Therapeutic Window, Narrow Therapeutic Index, and Target Concentration Strategies.
Answer
243
How is TDM typically performed, including sample types, lab techniques, and influencing factors?
Answer
244
Using Methotrexate and Warfarin as examples, explain why TDM is clinically indicated and what is monitored.
Answer
245
Apply TDM principles to IV antibiotics like Vancomycin and Gentamicin, including target levels, dosing strategies (intermittent vs continuous/once-daily), and considerations for renal impairment.
Answer
246
How is clinical decision-making guided by TDM results?
Answer
247
Summarize how emerging trends and technologies like pharmacogenomics, point-of-care testing, and AI are changing the landscape of personalised medicine.
Answer
248
Know the underpinning principles of the prescribing framework for healthcare professionals.
Answer
249
Outline how the perspectives, attributes, and attitudes of patients and prescribers influence prescribing decisions.
Answer
250
Name some modifiable and non-modifiable influences on prescribing practice.
Answer
251
Explain how intersecting influences shape prescribing practice.
Answer
252
Discuss the importance of context and setting, using the prison setting as an example to illustrate unique influences and challenges in prescribing.
Answer
253
Define scope of practice and explain its relationship to scope of competence.
Answer
254
Identify the factors that determine an individual's scope of practice.
Answer
255
Suggest possible scopes of prescribing practice for newly qualified pharmacists and how their scope might expand.
Answer
256
Why is it important to evaluate our practice?
Answer
257
What are some structured tools and methods used for evaluating practice?
Answer
258
When should we evaluate our practice?
Answer
259
Describe the types of concerns that the GPhC can investigate regarding fitness to practise.
Answer
260
Outline the four stages the GPhC follows for managing concerns.
Answer
261
Explain the two limbs of the Real Prospect Test applied by the Investigating Committee.
Answer
262
What are the possible outcomes from the GPhC's Investigating Committee and Fitness to Practise Committee?
Answer
263
Describe the actions that should be taken following a dispensing error.
Answer
264
What are the key responsibilities of Safeguarding Adult Boards?
Answer
265
List some relevant legislation related to safeguarding.
Answer
266
Name the different types of abuse recognized in safeguarding.
Answer
267
What actions should be taken when you are concerned about a vulnerable adult?
Answer
268
Describe the key principles to follow when managing safeguarding concerns, including what to avoid.
Answer
269
What is the process for a safeguarding referral from community pharmacy or a GP?
Answer
270
What steps are taken if the at-risk adult denies all abuse?
Answer
271
Explain how causality of ADRs is assessed, mentioning the TRIP acronym.
Answer
272
What are some strategies for preventing ADRs?
Answer
273
How are ADRs managed?
Answer
274
Describe the role and scope of the Yellow Card scheme.
Answer
275
What types of suspected adverse events should be reported to the Yellow Card scheme?
Answer
276
What are the main consequences of polypharmacy?
Answer
277
What are some solutions or interventions for managing polypharmacy, including deprescribing tools and the person-centred approach?
Answer
278
Describe the different models and phenotypes of Frailty.
Answer
279
What are the key assessment tools for frailty, such as the Comprehensive Geriatric Assessment (CGA) and the G8 Screening Tool?
Answer
280
Explain the components and process of a CGA.
Answer
281
List the components of the G8 screening tool.
Answer
282
Describe the common bone conditions found in the elderly.
Answer
283
Explain the process of bone ageing and pathophysiology.
Answer
284
What are the modifiable and non-modifiable risk factors for poor bone health?
Answer
285
How is Bone Health linked to Cancer and its treatments?
Answer
286
What are some Fall Risk Assessment Tools, their risk factors, and who should be assessed?
Answer
287
How is poor bone health diagnosed and screened for, including the use of DXA scans and lab tests?
Answer
288
What are the key Falls Prevention Strategies (Exercise, Lifestyle, Nutrition)?
Answer
289
How is Frailty managed?
Answer
290
What are the first-line and alternative treatments for poor bone health?
Answer
291
Explain the importance of co-management and starting osteoporosis treatment in post-fracture care.
Answer
292
What is involved in the monitoring and follow-up of patients receiving treatment for poor bone health?
Answer