• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/173

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

173 Cards in this Set

  • Front
  • Back
What cell cycle phase(s) do most anti-cancer agents target?
S-phase, because this is where all proliferation occurs
What class of drug is Mechlorethamine?
Alkylating Agents
MOA of Alkylating Agents
Crosslinking of DNA, and Alkylation (or carbamoylation) of DNA and/or protein

The end result is DNA damage
Common Property of Alkylating agents
* They become strong electrophiles through formation of carbonium ion intermediates, or transition complex with target molecules
* This reaction leads to formation of covalent bond by alkylation of nucleophilic moieties (phosphate, amino, sulfhydryl, hydroxyl, carboxyl, imidazole groups)
Phosphoramide Mustard
Active metabolite of Cyclophosphamide (also toxic)
Mechanism of Resistance of Alkylating Agents
* Acquired resistance is a common event; resistance to one agent often imparts cross resistance to others.
* Decreased permeation of actively transported drugs.
* Increased activity of DNA repair enzymes.
* Increased production of nucleophilic substances, principally thiols such as glutathione, that can conjugate with and detoxify electrophilic intermediates.
* Alternative: Methods are used to deplete glutathione, e.g. a sulfhydryl compound, BSO, buthionine sulfoximine.
* Increased rates of metabolism of activated forms of cyclophosphamide to its inactive metabolites.
Toxicities of Alkylating Agents
* As a class, highly leukemogenic.
* Cumulative bone marrow suppression.
* Toxic (a lesser extent) to intestinal mucosa.
* Pulmonary fibrosis.
* At high-dose regimen, endothelial damage.
Examples of Alkylating Agents
* Nitrogen Mustards –Mechlorethamine, Cyclophosphamide, Chlorambucil

* Nitrosourea - Carmustine

* Alkyl sulfonate - Busulfan

* Triazenes – Dacarbazine

* Platinum Compounds – Cisplatin, Carboplatin, Oxaliplatin
What are the sub-classes of Alkylating agents, how are they classified, examples of each?
Two classes of agents in terms of their effect on DNA:

1. Intrinsically Reactive – active by itself or direct acting
* Mechlorethamine
* Carmustine
* Busulfan
* Chlorambucil
* Cisplatin
* Carboplatin
* Oxaliplatin

2. Require Metabolic Activation to alkylating species
* Cyclophosphamide
Mechanisms of resistance for
1. Impaired transport into cells
2. Impaired polyglutamate formation
3. (MAJOR) Increased of altered DHFR
4. decreased thymidylate synthase (b/c much lower levels of DHFR in the absence of this enzyme)
Examples of Antimetabolites
* Folic Acid and Analogs
- Methotrexate
- Pemetrexed

* Pyrimidine Analogs
- Fluorouracil (5-Fluorouracil, 5-FU),
- 5-FU analog Capecitabine
Cytarabine (Cytosine Arabinoside, AraC)
- Gemcitabine

* Purine Analogs
- Mercaptopurine

* Others
- Fludarabine Phosphate – Fludarabine
- Hydroxyurea
Antimetabolites interfere with DNA synthesis – means which phase of cell cycle arrest?
S
Which drug is the most important 5-FU analog; a drug with proven activity against colon and breast cancers.
Capecitabine
Which 5-FU analog is a drug with proven activity against colon and breast cancers?
Capecitabine
What kind of tumor is most susseptible to Capecitabine?
Tumors with elevated thymidine phosphorylase activity seem particularly susceptible to this drug.
How is Capecitabine activated?
Converted to 5’-deoxy-5-fluoro-cytidine by carboxylesterase in liver (and other normal & malignant tissues) followed by to 5’-deoxy-fluorodeoxyuridine by cytidine deaminase. Final activation occurs when thymidine phosphorylase cleaves off 5’-deoxy sugar, leaving intracellular 5-FU.
Mechanism of Resistance of 5-FU Analog Capecitabine
Loss or decrease in activity of enzymes necessary for activation of 5-FU to F-dUMP.
Amplification of thymidylate synthase, and altered thymidylate synthase that is not inhibited by F-dUMP.
Toxicities of 5-FU Analog Capecitabine
Toxicities include: severe diarrhea, and toxic effects similar to other chemotherapeutic agents against cancer.
What is Cytarabine(Cytosine Arabinoside; AraC) used for?
Most important antimetabolite used in therapy of acute myelocytic leukemia.

Single most effective agent for induction of remission in this disease.
What is the MOA of Cytarabine(Cytosine Arabinoside; AraC)?
Inhibition of DNA through Inhibition DNA polymerase and DNA elongation.
What is Gemcitabine(2,2-difluorodeoxy cytidine, dFdC) used for?
Most important antimetabolite in clinic in recent years, and part of first-line regimen for patients with metastatic pancreatic and non-small cell lung cancers.
Gemcitabine Mechanism of Action
Mechanism of action is similar to AraC, however, efficacy is not confined to S-phase; it is equally effective against confluent and growing cells.
Primary toxicity of Gemcitabine
myelosuppressive, others include flu-like syndrome.
Alkylating Agents
* Nitrogen Mustards
- Mechlorethamine
- Cyclophosphamide
- Chlorambucil

* Nitrosourea
- Carmustine

* Alkyl sulfonate
- Busulfan

* Triazenes
– Dacarbazine

* Platinum Compounds
– Cisplatin
- Carboplatin
- Oxaliplatin
Antimetabolites
* Folic Acid and Analogs
- Methotrexate
- Pemetrexed

* Pyrimidine Analogs
- Fluorouracil (5-Fluorouracil, 5-FU),
- 5-FU analog Capecitabine
- Cytarabine (Cytosine Arabinoside, AraC)
- Gemcitabine

* Purine Analogs
- Mercaptopurine

* Others
- Fludarabine Phosphate
- Fludarabine Hydroxyurea
Microtubule (Mitotic Spindle) Agents
* Vinca Alkaloids
– Vinblastin
- Vincristine
- Vinorelbine

*Others:
- Paclitaxel (Taxol)
- Docetaxel (Taxotere) – Promote polymerization of microtubules.
Topoisomerase Targeting Agents
* Etoposide
* Teniposide

* Camptothecin and analogs (Topotecan and Irinotecan)
Antibiotics
* Dactinomycin (Actinomycin D)
* Doxorubicin and Analogs
* Mitoxantrone
* Bleomycin
The Altered Phenotypic Traits of Tumor Tissue
1. Resistance to anti-growth signal (TGFbeta downregulation, Rb mutation, C-m yc overexpression)
2. Resistance to apoptosis (NF-kapaB activation, p53 mutation)
3. Invasion and metastasis (loss of cell-cell adhesion eg. E-cadherin inactivation, increased MMP secretion)
4. Inflammation (Cox-2 overexpression, cytokine production)
5. Increased angiogenesis (secretion of VEGF, FGF)
6. Lack of senscence (upregulation of telomerase)
7. Constitutive mitotic signaling (E GFR, Ras activation)
What are six leading cancer types in the United States?
1. Lung
2. Prostate
3. Breast
4. Colon
5. Lymphoma
6. Urinary bladder
What are the principles and benefits of combination chemotherapy?
Principles:
- Choose non-overlapping mechanism of action of the drug.
- Choose drugs with the least overlapping major toxicities.

Benefit(s):
- Combination gives better activity than single agent, because there would be side effects of combined treatments.
True or False?

In addition to growing cancer cells, normal cells are damaged by anti-cancer drugs.
True

Similar to growing cancer cells, normal tissue that proliferate (bone marrow, hair follicle, intestinal epithelium) are damaged by anti-cancer drugs, limiting their use.
What are the main sub-classes of Alkylating agents?
1. Intrinsically Reactive – active by itself or direct acting
- Mechlorethamine
- Carmustine
- Busulfan
- Chlorambucil
- Cisplatin, Carboplatin, Oxaliplatin

2. Require Metabolic Activation to alkylating species
- Cyclophosphamide
Mechanism of Action of Carmustine
The degreadation of carmustine (BCNU) causes generation of alkylating and carbamoyling intermediates which pind to DNA and other proteins
Mechanism of Action of Cyclophosphamide
picture slide
Mechanism of Resistance of Alkylating Agents
* Acquired resistance is a common event; resistance to one agent often imparts cross resistance to others. * Decreased permeation of actively transported drugs. * Increased activity of DNA repair enzymes. * Increased production of nucleophilic substances, principally thiols such as glutathione, that can conjugate with and detoxify electrophilic intermediates. Alternative: Methods are used to deplete glutathione, e.g. a sulfhydryl compound, BSO, buthionine sulfoximine. * Increased rates of metabolism of activated forms of cyclophosphamide to its inactive metabolites.
Toxicities of Alkylating Agents
* As a class, highly leukemogenic. * Cumulative bone marrow suppression. * Toxic (a lesser extent) to intestinal mucosa. * Pulmonary fibrosis. * At high-dose regimen, endothelial damage.
How are platinum compounds different from other Alkylating Agents?
* Exact mechanism(s) of action not known, but they form DNA adducts. * Major resistance is via DNA repair
What is Cisplatin used for?
broad activity, specifically useful in epithelial malignancies.
What major toxicities are associated with Cisplatin?
Major toxicities include renal proximal tubular necrosis, ototoxicity.
What is Carboplatin used for?
Ovarian cancer
What major toxicities are associated with Carboplatin?
Major toxicities include renal proximal tubular necrosis, ototoxicity. Similar to Cisplatin but less prevalent.
What is Oxaliplatin used for?
A wide range of cancers including ovarian and cervical cancers. Generally more effective than Cisplatin or Carboplatin because DNA repair enzyme proficient cells are equally effective.
"
Methotrexate
What event lead to the development of anti-folate analogs that specifically target folate-dependent enzyme targets of methotrexate?
A finding that methotrexate, an inhibitor of dihydrofolate reductase (DHFR), also inhibits folate dependent enzyme of purine and thymidine synthesis
Which agent is a Multitargeted Antifolate (MTA)?
"Pemetrexed, an important new folate analong which inhibits thymidylate and purine biosynthesis, in addition to dihydrofolate reductase (DHFR) inhibition.
What cancers has Pemetrexed shown activity against?
colon cancer, mesothelioma, and non-small cell lung cancer.
What are the two important enzymes targeted by folate analogs?
Thymidylate synthase and dihydrofolate reductase
Which folate analogs block the regeneration of tetrahydrofolate?
aminopterin and methotrexate
What is the MOA of folate analogues?
"* To function as a cofactor in one-carbon transfer, folate (dihydrofolate) is reduced by DHFR to tetrahydrofolate (FH4). * Single-carbon fragments are added enzymatically to FH4 (for example, conversion from serine to glycine), leading to 5,10-methylene FH4. * dUMP is converted to dTMP where thymidylate synthase transfers one carbon group to dUMP from methylene FH4 converting it to FH2. * DHFR is primary site of action for most folate analogs. Its inhibition leads to toxic effects via depletion of FH4 required for synthesis of purines and thymidylate (dTMP), precursors of DNA/RNA synthesis. This causes interruption of DNA/RNA synthesis. * At the same time, it causes vast accumulation of toxic substrate FH2 polyglutamate.
What are the major mechanisms of resistance to Methotrexate?
1. Impaired transport of drug into cells 2. Impaired polyglutamate formation 3. (MAJOR) increased or altered dihydrofolate reductace 4. decreased thymidylate synthase
What are the unique toxicities of methotrexate?
* Toxic against rapidly growing normal cells of bone marrow and GI epithelium. * Hepatotoxicity: fibrosis and cirrhosis * Nephrotoxicity due to drug precipitation
What is the MOA of Pyrimidine Analogs 5-FU?
* Inhibit biosynthesis of pyrimidine nucleotides. * Mimic these natural metabolites to such an extent that they interfere with the synthesis or function of nucleic acids. * The best characterized agents in this class are halogenated pyrimidines: fluorouracil and 5-fluorouracil (5-FU) * By several activation pathways, fluorouracil is converted in vivo to fluorodeoxyuridylate (F-dUMP) that irreversibly inhibits thymidylate synthase after acting as a normal substrate. * This inhibits the methylation of dUMP.
How does F-dUMP inhibit methylation of dUMP?
"* A –SH group of enzyme adds to C-6 of F-dUMP. Methylene FH4 then adds to C-5 of this intermediate. * In case of dUMP, a hydride ion of folate is subsequently shifted to methylene gr, and a proton is taken away from C-5 of bound nucleotide. * However, F+ cannot be abstracted from F-dUMP by enzyme, and so catalysis is blocked at the stage of covalent complex formed by F-dUMP, methylene FH4 and –SH of enzyme.
Does Cytarabine (Cytosine Arabinoside; AraC) need to be activated?
Yes, must be activated to 5’-mono-phosphate nucleotide (AraCMP)
How is Cytarabine (Cytosine Arabinoside; AraC) activated?
deoxycytidine kinase activates to 5’-mono-phosphate nucleotide (AraCMP)
Mechanism of Resistance of Cytarabine
"* Increased cytidine deaminase, a degradation enzyme that deaminates AraC to non-toxic metabolite, arauridine. * Decreased levels of deoxycytidine kinase that converts AraC to AraCMP. * A second degradation enzyme dCMP deaminase, converts AraCMP to inactive metabolite AraUMP.
Primary toxicities of Cytarabine
myelo-suppression and others include GI disturbances.
What are Mercaptopurine and 6-Mercaptopurine (6-MP) used for?
"* Useful in the treatment of malignant diseases. * Also used as immunosuppressive and anti-viral agent.
What is the MOA of Mercaptopurine and 6-Mercaptopurine (6-MP)?
* Inhibits multiple pathways in conversion of inosine monophosphate (IMP) to adenine and guanine nucleotides. * Must be activated to nucleotide form by hypoxanthine guanine phosphoribosyl transferase (HGPRT).
Mechanism of Resistance for Mercaptopurine and 6-Mercaptopurine (6-MP)?
"* Acquired resistance – deficiency or complete lack of HGPRT. * Decreased affinity of enzyme for substrate. * Others include: decreased drug transport, increased rate of degradation of drug, increased activity of MDR protein.
What are toxicities of Mercaptopurine and 6-Mercaptopurine (6-MP)?
hyperuremia, bone marrow suppression, nausea, vomiting, jaundice.
What is Fludarabine Phosphate Fludarabine used for?
"Active in chronic lymphocytic leukemis and low-grade lymphomas.
What is the MOA of Fludarabine Phosphate Fludarabine?
* After rapid dephosphorylation to nucleoside fludarabine by membrane 5’-ectonucleotidase, it is rephosphorylated intracellularly by deoxycytidine kinase to active triphosphate. * Inhibits DNA polymerase etc, and is incorporated into DNA and RNA. * Although the exact mechanism of activity is not well understood, it causes DNA chain termination and induction of apoptosis.
What are toxicities of Fludarabine Phosphate Fludarabine?
myelosuppression, nausea and vomiting, and chills and fever.
What is the MOA of Hydroxyurea?
* Inhibits ribonucleoside diphosphate reductase, an enzyme that catalyzes reductive conversion of ribonucleotides to deoxyribonucleotides; a crucial and a rate limiting step in the biosynthesis of DNA. * Specific for S-phase.
What is the major mechanism of resistance for Hydroxyurea?
elevation of reductase activity
What are toxicities of Hydroxyurea?
hematopoietic depression, GI disturbances and mild dermatological reactions.
What is oncogene addiction and how does oncogene addiction relate to intracellular signaling?
Tumor cells become ‘addicted’ to that growth stimulatory pathway constitutively activated as a result of protooncogene or tumor suppressor gene mutation. Thus, if a cancer cell has dozens of mutations associated with cell division but they are primarily using the mutated Kras pathway to provide signals that allow them to grow, then inhibiting the Kras pathway with various drugs will interfere with cancer growth. This is true even though several other pathways could theoretically be used by the cell. In contrast, normal cells are not addicted to any particular pathway.
Why is it important to diagnose cancer early?
If tumors could be detected very early, before their inherent genetic instability allows them to make genetically variant daughter cells that are resistant to drugs or better at metastasizing, this would greatly enhance cancer prevention and therapy
What are the differences between benign and malignant tumors?
Growth rate
- Benign
* low mitotic rate
* normal mitoses
* normal nucleoli
- Malignant
* high mitotic rate
* abnormal mitoses
* large nucleoli
Differentiation
- Benign
* resembles normal
* maintains normal functions
- Malignant
* often poor
* lost or altered function
Spread
- Benign
* encapsulated
* no invasion
* no metastases
- Malignant
* no capsule
* locally invasive
* matastases common
How are tumors named?
By tissue type and Behavior:
- Benign = -oma
- Malignant = ...carcinoma or ...sarcoma
Papilloma
Benign tumor in the stratified squamous epithelium

or

Benign tumor in the transitional epithelium
Squamour cell carcinoma
Malignant tumor in the stratified squamous epithelium
Adenoma
Benign tumor in a glandular epithelium
adenocarcinoma
Malignant tumor in a glandular epithelium
transitional cell carcinoma
Malignant tumor in the transitional epithelium
Fibroma
Benign non-endothelium fibrous tissue tumor
fibrosarcoma
Malignant non-endothelium fibrous tissue tumor
lipoma
Benign non-endothelium fatty tumor
liposarcoma
Malignant non-endothelium fatty tumor
leiomyoma
Benign non-endothelium smooth muscle tumor
leiomyosarcoma
Malignant non-epithelial smooth muscle tumor
rhabdomyoma
Benign non-epithelial striated muscle tumor
rhabdomyosarcoma
Malignant non-epithelial striated muscle tumor
synovioma
Benign non-epithelial synovium tumor
synovial sarcoma
Malignant non-epithelial synovium tumor
chondroma
Benign non-epithelial cartilage tumor
chondrosarcoma
Malignant non-epithelial cartilage tumor
osteoma
Benign non-epithelial bone tumor
osteosarcoma
Malignant non-epithelial bone tumor
haemangioma
Benign non-epithelial blood vessel tumor
angiosarcoma
Malignant non-epithelial blood vessel tumor
benign taratoma
Benign non-epithelial germ cell tumor
malignant teratoma
Malignant non-epithelial germ cell tumor
naevus
Benign non-epithelial neuroectoderm tumor
melanoma
Malignant non-epithelial neuroectoderm tumor
What is the evidence that tumors change and evolve with time?
the growth of cancer compares to fetal growth, how neoplastic growth results from disturbance of the normal balance between proliferation and apoptosis, genes involved in the cell cycle.
What causes cancer? How do they know?
Environmental factors coupled with genetic succeptibility cause cancer. Remember "Cappadocia, mesothelioma
villages"
What is the evidence that smoking causes cancer?
PAH and NNK are metabolically activated which leads to DNA adducts. This in tern leads to mutations in k-RAS, p-53 and other critical genes.
How does the tumor microenvironment/stroma affect tumor cells?
Tumor cells exist in an intimate cross-signaling environment with stromal (mesenchymal) cells, these include macrophages which can affect tumor growth in many ways.
What is the difference between innate and adaptive immune cells?
Innate immune cells: neutrophils, macrophages, dendritic cells and natural killer cells

Adaptive immune cells: T-cells and B-cells
What is the significance of tumor angiogenesis?
Like all tissues, a tumor cannot survive or grow without a blood supply. Tumor cells release factors that stimulate new blood vessel growth. In normal tissues, epithelial cells secrete inhibitors of endothelial cell growth, and vice-versa. As the tumor progresses (becomes more advanced), the nature of these signals changes and each cell type stimulates the growth of the other.
How does tumor angiogenesis relate to new drug discovery?
Anti-angiogenic drugs are being applied along with conventional cytotoxic agents. The great advantage to their use is that endothelial cells are genetically stable, like all normal cells, and won’t mutate to become resistant to these drugs.
What is EMT (epithelial-mesenchymal transition)? How does this relate to metastasis? What are the stages of metastasis?
. Once there, tumor cells undergo MET (mesenchymal-epithelial transition) to become more epithelial-like as they grow into a new tumor at another body site. This tumor will usually be recognizable as having been derived from the original primary tumor, but will be more advanced (and cellularly heterogeneous and thus harder to kill) than the primary tumors. Platelets aggregate around tumor cells in the circulation and protect them as a physical barrier from being killed. The platelets also secrete factors that enhance tumor cell survival. Each type of cancer has characteristic organ sites they metastasize to, but within these trends show great individual variation.
What is the mechanism of cachexia?
Tumors release TNFa, which in large concentrations can cause cachexia (a general weakening and appetite loss) and a sepsis-like condition.
Why are cancer patients more sensitive to infection?
* Neutropenia
* Abnormal microbial flora
* Defective cell-mediated immunity
* Defective humoral immunity
* Malnutrition
* Splenectomy
* Impaired monocyte-macrophage system
* Impaired chemotaxis, phagocytosis, bactericidal activity
* Broken physical defence barriers
How do tumors escape immune surveillance?
White blood cells infiltrate into tumors, including macrophages. These are recruited from the circulation and secrete many factors that stimulate tumor growth, depress tumor apoptosis, and cripple the adaptive immune system so the body’s natural defenses are inadequate to fight the disease. Therapies are aimed at re-activating macrophages such that they again become anti-tumor (can kill tumor cells) as they were in cancer-free people and at very early stages of tumor growth are being used along with conventional cytotoxic treatments.
How prevalent is cancer in teh US?
2nd most common cause of death in US
 1,479,350 new diagnoses in 2009
 562,340 deaths
Define chemoprevention
Chemoprevention –the use of specific natural or synthetic agents to reverse, suppress or prevent the carcinogenic process, thereby preventing the development of clinically evident cancer
Define Primary chemoprevention patient
Primary –general population of healthy individuals at risk
Define Secondary chemoprevention patient
Secondary –individuals with premalignant lesions to prevent progression to cancer
Define Tertiary chemoprevention patient
Tertiary –prevention of a second primary cancer
Define early detection
Early detection –of a premalignant or early stage cancer
Breast Cancer Chemoprevention
 192,370 new diagnoses
 40,170 deaths
 70-85% of sporadic breast cancers are ER+
 Chemoprevention is aimed at altering ER stimulation
How is it determined who should receive Breast Cancer Chemoprevention?
Gail Model
 Age
 Age at menarche
 Age at first live birth or nulliparity
 Number of breast biopsies
 History of atypical hyperplasia
 Number of first-degree relatives with breast cancer
 Race/Ethnicity
Who is included in the Primary Chemoprevention Population for breast cancer?
 Genetic mutations
 First degree relatives with breast or ovarian
 History of thoracic irradiation
 Gail model 5-year risk ≥1.7%
Who is included in the Secondary Chemoprevention Population for breast cancer?
Lobular carcinoma in situ (LCIS)
Chemoprevention for Women at high risk for breast cancer
 Mastecomyor bilateral oophrectomy
 Only recommended for those with LCIS, compelling family history or known BRCA mutation
 Can decrease risk 90-95%
 Pharmacological prevention: SERMs
 Recommended for most high risk women ≥35yo
 Contraindicated in women with h/x of thrombosis, TIA or potential pregnancy
 Tamoxifen
 Raloxifene
Tamoxifen (breast cancer)
 FDA approved for risk reduction of breast cancer in those at high risk or with ductalcarcinoma in situ (DCIS)
 20mg daily for 5 years
 Pre-and post-menopausal
Raloxifene (breast cancer)
 FDA approved for risk reduction of invasive breast cancer in post-menopausal women at high risk or with osteoporosis
 60mg daily for 5 years
 Post-menopausal ONLY
What are the Pros for using SERMs as chemoprevention in breast cancer?
 Increase bone mineral density
 Reduce LDL and total cholesterol
What are the Cons for using SERMs as chemoprevention in breast cancer?
 Tamoxifen increases endometrial proliferation
 2x risk of endometrial cancer
 Thrombosis
 May increase pro-coagulant production in liver
 Not for women with h/o DVT or PE, on anticoagulant therapy, smokers
 Should be held 2-4 weeks prior to surgeries
 Cataracts
 Tamoxifen > Raloxifene
 Not if pregnant, planning to become pregnant or breastfeeding
Which SERM requires metabolism of active metabolites through CYP3A4, 2C9 and 2D6?
Tamoxifen
Which SERM has an active parent compound and is affected by hepatotoxicity?
Raoxifene
PROS and CONS of Tamoxifen
 Pros
 Effective
 Lots of data on long-term use
 Osteoporosis prevention (post-menopausal only)
 Can be used in pre-menopausal women
 Cons
 Menopausal sxs
 DDI
 2D6 inhibitors (SSRIs)
 Warfarin
 3A4 and 2C9 inhibitors/inducers
 Cost: $100/month x12 months x5 years= $6000
PROS and CONS of Raloxifene
 Pros
 Effective
 Osteoporosis prevention (FDA)
 Less risk of thrombosis, endometrial cancer, cataracts
 Cons
 Weight gain
 Menopausal symptoms
 Sexual dysfunction slightly worse (STAR)
 DDI
 Cholestyramine
 Cost: $123/month x12 months x5 years= $7380
Prostate Cancer Chemoprevention
 3rd most common cancer worldwide
 146,970 new diagnoses in US in 2009
 49,920 deaths in US in 2009
 Chemoprevention aimed at decreasing androgen stimulation of prostate tissue
Who should receive Prostate Cancer Chemoprevention?
 Primary
 Patient-specific
 Risk assessment
 Elevated PSA levels
 Rapid PSA velocity
 Sub-Saharan African ethnicity
 Family history
 Secondary
 High-grade prostate epithelial neoplasia(HGPIN)
Prostate Cancer Chemoprevention for high-risk men
 Digital Rectal Exam and PSA starting at 40yo
 Subsequent screening depending on results
 Some controversy over when to pursue further testing
 5αreductase inhibitors
 Finasteride
 Dutasteride
MOA of 5α reductase inhibitors
Inhibit 5α reductase inhibitors so testosterone cannot be converted to DHT
Pharmacological chemoprevention for prostate cancer
 Finasteride
 5mg once daily x7 years
 Dutasteride
 0.5mg once daily x4 years
 Not FDA-approved
 Significant increase in high-grade, aggressive tumors compared to placebo (37% v 22%)
 Patients did not have more extensive disease
 Reanalysis showed no increased risk
NEJM 2003; 349(3):
REDUCE trial
 8231 men 50-75yo
 Primary endpoint: prevalence of cancer on prostate biopsies at 2 & 4 years
 Preliminary results
 Dutasteride reduced risk by 23% at 4 years
 No increase in high-grade, aggressive disease
ADRs associated with 5α reductase inhibitors
 ADE
 Sexual dysfunction
 Impotence, decreased libido, ejaculation disturbances
 Gynecomastia
 Does not decrease with continued use
 Caution with liver disease
 Pregnant women or those trying to conceive should NOT handle
 Wear gloves
PROS and CONS of Finasteride use for chemoprevention of prostate cancer
 Pros
 Effective
 Cons
 May increase risk of high-grade tumors
 ADE
 Muscle weakness
 Dizziness, postural hypotension
PROS and CONS of Dutasteride use for chemoprevention of prostate cancer
 Pros
 Lower incidence of sexual dysfunction
 Inhibits both isoformsof 5AR
 Cons
 May increase TSH
 Awaiting full results
Prostate Cancer: Chemoprevention, or not?
* Pros
 Reduction in risk
 Improves sxs of BPH
 Better early detection
 Decreases HGPIN

*Cons
 No proven effect on mortality
 Unsure if increases risk of high-grade tumors
 Sexual side effects
 Cost
Colorectal Chemoprevention
 3rd most common cancer worldwide
 146,970 new diagnoses in US in 2009
 49,920 deaths in US in 2009
 Normal colon leads to polyp leads to cancer
 Chemoprevention aimed at reducing polyp formation and development
Colorectal Cancer Cehmoprevention: ASA/NSAIDs/COX-2 inhibitors
 Epidemiological data
 Aspirin, NSAIDs and COX-2 inhibitors may prevent polyp formation
 Studied in patients with familial syndromes
 4-6 aspirin per week can decrease incidence by 30-50%
Why not give everyone aspirin?
* Pros
 Effective
 ASA cardioprotective
 Consider in patients with familial syndromes or personal h/o

*Cons
 Benefit may take 10-20 years
 Benefit not maintained after d/c
 Higher doses
 GI bleed risk
 Increased cardiovascular risk
Cervical Cancer Chemoprevention
 11,270 women in the United States/yr
 4,070 will die this year
 Routine Pap smears decrease morbidity/mortality in developed countries
 90% detection
 Cervical cancer is the second most common cancer among women worldwide
 83% of cases occurring in developing countries
Discuss Cervical Cancer Risk Factor: Human Papillomavirus(HPV) Infection
 Worldwide prevalence ~ 10%
 Small DNA viruses
 Role in cervical cancer
 Large link to viral oncogenes E6 and E7
 Effects cell cycle/growth arrest signals
 Types 16 and 18 implicated
Chemoprevention fo Cedrvical cancer
Vaccination with Gardasil
• QuadrivalentHPV vaccine (types 6, 11, 16, 18)
• Indicated in females
– Age 9-26
• Administration
– 3 injections over 6 months
– ~$400/series
Gardasil
 Efficacy
 ~95% of women protected against types 16/18
 >98% retain seroconversion at 5.5 years
 Relatively safe
 Injection site reactions
 Some anaphylactic reactions have occurred
 Two cases of Guillain-Barre syndrome reported
Gardasil Pearls
 Does not treat cervical cancer or genital warts
 Not to use in pregnant women
 Do not use if severely allergic to yeast
 Must continue routine screening
 Duration of action? unknown
Diet/Supplements for Chemoprevention
 Vitamin A for lung
 Vitamin E/selenium for prostate
 Vitamin D/calcium for colorectal
 Diet/alcohol consumption
Vitamin A (Retinoids)
 Multiple dietary sources
 Antioxidant
 Epidemiological studies suggest may protect against development of lung cancer in smokers
Trials involving Vitamin A (Retinoids) and the bottom line
 ATBC trial
 Men who took beta-carotene had 18% increased risk of lung cancer, 8% increased mortality risk
 Follow-up period of 8 years: Men in beta-carotene group with 7% increased mortality
 CARET
 28 percent more lung cancers were diagnosed and 17 percent more deaths occurred after 4 years of supplementation
 Associated with increased risk of lung cancer in smokers
Selenium and Vitamin E

Found in plants grown in Se-rich soil, meat

Activation of DNA repair, p53 tumor suppressor gene

Antioxidant

Found in plants grown in Se-rich soil, meat

Activation of DNA repair, p53 tumor suppressor gene
What was the outcome of the SELECT trial?
No difference between placebo, vit E, slelenium, and vit E and selenium combo therapy
Calcium and Vitamin D
 Calcium
 Binds fatty acids and bile in gut lumen, inhibits gut mucosal proliferation
 Prostate
 Ca >2gm/day associated with increased risk
 Vitamin D
 Induction of apoptosis, inhibition of cell proliferation, modulates calcium absorption
 Increased intake may be associated with a decrease in colorectal cancer, other GI tumors
NIH-AARP Diet and Health Study
 >492K US men and women aged 50-71yo
 Dietary Ca ≥1300mg/day associated with decreased overall cancer risk in women
 Higher dairy food intake associated with decreased risk of digestive tract cancers
 Higher calciumintake associated with decreased risk of colorectal cancer
Fiber
 Increased fecal volume, dilution of carcinogens due to increased bulk, faster transit time and stimulation of gut flora decreases exposure to carcinogens
 Colorectal cancer
 Overall inconsistent benefit
 EPIC suggested up to 40% decrease compared to low-fiber diets
 Breast cancer
 Difficult to dissociate from impact of fat in diet
Dietary Fat
 Association between animal fat and increased risk of breast, colorectal cancer
 Closely linked to obesity, other poor dietary choices
Alcohol Consumption
 Million Women Study (the UK)
 1.28 million middle-aged women
 Studied moderate alcohol consumption
Alcohol and Cancer Risk
Additional Cancer Risk/1000 Women with1 Additional Drink/Day
Overall
15
Breast
11
Oropharyngeal
1
Rectal
1
Esophageal/Laryngeal
0.7
Hepatocellular
0.7
Cause Cancer?
 Grilled foods
 Carcinogens are generated during charring
 Artificial sweeteners
 Laboratory data suggest aspartame and saccharin can be carcinogenic in rats
 Fluoride
 Increase in osteosarcomain rats
In a Nutshell…what are the nutritional and supplement recommendations?
 Eat a well-balanced diet
 Obtain most food from plant sources
 Maintain a healthy weight
 Choose whole grains
 Limit processed foods

 And now…
 Protect your bones!
 Limit alcohol intake
Cancer Prevention Screening Goals
 Increase detection of precancerous lesions
 Detect cancers prior to spread
Skin Cancer Screening
 Most common cancer diagnoses in US
 8,650 deaths in US in 2009
 US lifetime risk: 1 in 55 adults
Types of Skin Cancer
Squamous Cell
Basal Cell
Melanoma
Risk Factors for Melanoma
 Many nevi, irregular nevi, large nevi
 Family history
 Places with intense, year-round sunshine
 UV radiation increases up to 5% for every 1000 feet above sea level
 Fair skin
 Inability to tan
 Sun exposure
 Severe sunburns as a child
Screening for Melanoma
 Asymmetry
 Border irregularity
 Color variegation
 Diameter >6mm
 Evolution
Skin Self-Exam Technique
 Performed monthly
 Head to toe
 Scalp
 Include non sun-exposed areas
 Underarms
 Between fingers & toes, nail beds
 Report any suspicious nevi to physician
Skin cancer prevention
 Visual self-examination
 >90% of melanomas recognized by naked eye
 Cover up
 Long sleeves, wide-brimmed hat
 UV-blocking sunglasses to protect eyes
 Use a sunscreen SPF 15 or higher
 UVB protection
 Reapply q2 hours
 Limit intense sun exposure
 Tanning beds
Why Colorectal Cancer Screening
Stage at Diagnosis
5-year Survival
I
>90%
II
70-85%
III
24-59%
IV
<5%
Colorectal Cancer Screening
 Colonoscopy
 Flexible Sigmoidoscopy
 Fecal Occult Blood (FOBT)
 Fecal immunochemical test (FIT)
 Double-Contrast Barium Enema
 Virtual colonoscopy
 Molecular testing
 Stool DNA tests
When should screening begin?
 Beginning at age 50, both men and women should follow one of the following options:
 Colonoscopy every 10 years
 Yearly FOBT + Sigmoidoscopyevery 5 years*
 Sigmoidoscopyevery 5 years*
 Double-contrast barium enema every 5 years*
 High-risk patients should begin screening earlier with colonoscopy
 African Americans should begin at age 45
 If one 1stdegree or two 2nddegree relatives, begin at 40yo or 10 years before earliest diagnosis
 Repeat q 3-5 years
 If IBD, begin 8-10 years after symptom onset
 Repeat q 1-2 years
Colorectal Screening: why not?
 Bowel preps
 Concern over adequate analgesia
 Lack of perceived risk
 Embarrassment
 Access to care
Bowel Preps
 Oral gastrointestinal lavage solutions
 Polyethylene glycol (PEG)
 +/-bisacodyl
 Saline laxatives
 Sodium phosphate
 Caution in renal impairment
 Usually with bisacodyl
 Clear liquid diet 24h prior
Breast Cancer Screening
Stage at Diagnosis
5-yearSurvival
0-I
100%
II
86%
III
57%
IV
20%
Breast Cancer Screening Recommendations
NCCN Breast Cancer Screening and Diagnosis Guidelines v.1.2010
 Breast awareness
 All women starting at 20yo
 Clinical breast exam
 Q 1-2 years starting at 20yo
 Annually starting at 40yo
 Every 6-12 months if high-risk
 Annual mammogram
 All women starting at 40yo
 High-risk women starting at 35yo
 If familial history, starting at 25yo or 5-10 years before diagnosis
 Annual MRI
 High-risk women
Mammogram Controversy
 US Preventative Services Task Force
“The USPSTF recommends against routine screening mammography in women aged 40 to 49 years. The decision to start…should be an individual one and take patient context into account, including the patient’s values regarding specific benefits and harms.”
 Still recommended by American Cancer Society, National Cancer Institute and National Comprehensive Cancer Network