• 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

1/181

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;

181 Cards in this Set

  • Front
  • Back
What is primary cancer prevention?
Measures to ensure that cancer never develops
What is secondary cancer prevention?
Measures used to dectecting and treating cancer while in its earliest most curable stage.
What are socioeconomic factors? What are some examples?
Socioeconomic factors are the social and economic experiences that help mold one's attitude, personality, and lifestyle. Examples include: Poverty, unemployment, life style, low education level, and limited or no access to health care.
What is diversity?
differences in beliefs, standards, and ways of life need to be taken into account for patients in order to provide them with adequate healthcare.
What are cancer disparites?
adverse differences in cancer incidence (new cases), cancer prevelance (all existing cases), cancer death (mortality), cancer survivorship, and burden of cancer or related health conditions that exist among specific population groups in the U.S.
What is the leading predictor in cancer disparities?
Poverty
What are the top 3 causes of death in the US?
1. Cardiovascular disease
2. Cancer
3. Stroke/ diabetes
What is a neoplasm?
abnormal mass of cells typically exhibiting progressive and uncontrolled growth; classified by the cell type from which they originate and their biologic behavior.
Define neoplastic transformation
multistep process involving three sequences of events: initiation, promotion, and progression.
What is initiation?
the first step in turning a normal cell cancerous as by drugs, chemicals, biological or other agents that are capable of producing irreversible changes in the DNA of a cell.
What is promotion?
the second step, where agents or cocarcinogens cause unregulated accelerated growth in previously initiated cells. Examples include: hormones, plant products, chemicals, and drugs.
What is progression?
- tumor cells acquire malignant characteristics that include changes in growth rate, invasive potential, metastatic frequency, morphologic traits, and responsiveness to therapy.
What is the purpose of screening?
• Screening is done to detect pre-cancerous changes, or cancer at an early stage. It is usually preformed on people who are asymptomatic.
What are some concerns for screening?
overdiagnosis, & false positives.
What are some challanges for screening?
- main challenge is cost. Making cancer screening readily available to people at costs they can afford. Availability of cancer screening is another challnge. People living in the country don’t have access to lots of medial care, and some people work during times that most people offer for cancer screens.
What are the screening guidelines for breast cancer?
o Yearly mammogram for women starting at 40. (the age they should be stopped at should depend on the individual, by considering risks and benefits of screening compared to overall health status and longevity.)
o Clinical breast exams should be part of periodic health exams
o screening mammography reduces breast cancer deaths in women ages 40-74.
What are the screening guidlines for prostate cancer?
The ACS recommends that both the prostate-specific antigen blood test and the digital rectal examination be offered annually, beginning at age 50, to men who have a life expectancy of at least 10 years and to younger men who are at high risk.
What are the guidlines for colon and rectum?
Beginnign at age 50, men and women should follow one of the examination schedules :
A fecal occult blood test every year and a flexible sigmoidosscopy every 5 years
A colonoscopy every 10 years
A double-constrast barium enema every 5-10 years.
What are the guidelines for cancer-related checkup?
A cancer related checkup is recommended every 3 years for people aged 20-40 and every year for people age 40 and older.
What are the guidelines for uterus?
Cervix: All women who are or have been sexually active or who are 18 and older should have an annual Pap test and pelvic examination. After three or more consecutie satisfactory examinations with notmal findings, the pap test may be preformed less frequently
Endometrium: Women at high risk fro cancer of the uterus should have endometrial tissue examined when menopause begins.
What is the role of chemoprevention?
• The use of natural or laboratory-made substances to prevent a disease such as cancer. The regular use of aspirin is known to reduce the risk of the polyps from which colorectal cancer arises. This is an instance of chemoprevention.
Initiatives to decrese smoking prevalence should be targeted to which of the following groups?
a.high school graduates
b.high school drop outs
c. college graduates
d.persons with some college
B. high school drop outs
More than 75% of all prostate cancer occur in which of the following groups?
a. African American men
b. Men over the age of 65
c. Asian/Pacific Islanders
d. Men between the age of 45 and 50
B. Men over the age of 65
Neoplastic transformation in human cancers involves the following sequences fo events:
a.stimulus, response, and initiation
b. Risk, exposure, and progression
C.Initiation, promotion, and progression
D. Irritation, promotion, and susceptibility
C. Initiation, promotion, and progression
According to the American Cancer Society guidelines, for optimal breast health, asymptomatic women should do which of the following?
A. Annual breast self-examination (BSE) and mammography starting at age 50
B. clinical breast examination (CBE)and BSE monthly, starting at age 40
C. Monthly BSE and annual CBE and mammogram starting at age 40
D. Annual CBE and mammogram every 3 years starting at age 50
C. Monthly BSE adn annual CBE and mammogram starting at age 40
Poor survival statistics, delayed diagnosis, and increased mortality amoung minority populations stem from barriers related to:
a.Primary prevention and lack of access
b.Socioeconomic status adn secondary prevention
c.lack of access and illiteracy
d.secondary and tertiary prevention













Answer














Side 3














Character MapSave Flashcards Please note! You must enter both questions and answers to save permanently.





Copyright © 2001-2012 Tuolumne Technology Group, Inc.

API | News | About | Help | Terms of Service | Privacy Policy
A. primary prevention and lack of access
A 45- year- old African American man is seen by an advance practice nurse for a routine physical. Which of the following would be most appropriate screening test for his race and age?
a. DRE and PSA
b. Colonoscopy
c. Ultrasound of the bladder
d. stool for occult blood
a. DRE and PSA
Reducing cancer risk related to sexual life-styles can most realistically be accomplished by:
a.avoiding homosexuality
b.engaging in safe sexual practices
c. practice abstinence
d.avoiding intravenous drug use
b.engaging in sage sexual practices
The overall incidence of cancers can best be reduced by:
a.early detection
b.screening
c.prevention
d.early treatment
C.prevention
The American Cancer Society estimates that 80% of all cancers are associated with which of the following:
a.Genetic predisposition
b. Racial and ethnic factors
c. Socioeconomic factors
d. Environmental exposures
D. Environmental exposures
Healthy People 2010 focus areas identify cancer as the ____ area of focus.
a. 5th
b. 4th
c. 2nd
d. 3rd
d. 3rd
What is the role of the healthcare provider in patient diagnosis of cancer?
• Healthcare providers need to have:
o up to date knowledge
o high index of suspicion for abnormal findings
o Pay close attention to physical exam and pt’s history
o Be aware of American Cancer Society’s recommendation for early cancer detection in average-risk and asymptomatic people.
o Target elderly for early detection, screening and diagnostic efforts.
o Adequately assess functional status and comorbidities
o Have high index of suspicion based on known risk factors
o Guidance towards appropriate testing procedures for accurate diagnosis.
o Generate a team effort in assisting, diagnosing and treating patient. Should include: physician, nurse, oncology specialists, pathologists, and technicians.
o **Comfortable, open, and honest relationships with patients are best.***
What are tumor markers?
Hormones, enzymes or antigens produced by tumor cells and measurable in the blood of persons with malignancies. The usefulness of tumor marker in diagnosis and tracking of malignancies depends on the markers sensitivity and specificity.
What does the tumor marker PSA stand for and what does it diagnose?
Prostate specific antigen
Used to diagnose prostate cancer
What cancers does CEA diagnose and what does CEA stand for?
(carcinoembryonic antigen)- breast, colorectal, lung cancer
What does the tumor marker HCG detect and stand for?
Human chorionic gonadotropin)-germ cell tumors, pregnancy
What does the tumor marker CA-125 detect?
Advanced epithelial ovarian cancer
What types of diagnostic imaging is used in the detection of cancers?
X-rays, PET scans, CT scans, mammography, ultrasound, and MRI
Why is diagnostic imaging important in the staging and diagnosis of cancer?
Allows for a non-invasive way to approximate the size and extension of tumors in patients.
What is TNM staging?
(tumor, node, mets)- most common. Used in breast and colorectal staging.
What is FAB staging?
French-American-British)- hematologic malignancies, leukemia, multiple myeloma.
What is Durie-Salmon system?
incorporates information regarding renal fx.
What is clinical staging the the TNM system?
o Based on professional judgment and measurement of the primary tumor’s size, location in the body, and evidence of disease through physical examination.
What is pathological staging in the TNM system?
o Practice of examination of the tissue of interest both grossly & microscopically to evaluate its characteristics and make an assessment as to the aggressiveness of the malignant tumor. Usually done after the tumor is surgically removed.
What is cytogenetics?
the use of molecular biologic testing to observe gene translocations and rearrangements.
Includes: Chromosome banding, FISH, and polymerase chain reaction.
How is a tissue biopsy used to diagnose and stage cancer?
o Definitive method for diagnosis of malignancy.
o Surgical procedure that involves obtaining a portion of tissue or excising the entire target tissue. Tissue obtained is then examined and subjected to diagnostic testing by the pathologist.
o Can determine malignant or benign
o Characteristics of malignant tissue can be examined to delineate tumor grade, aggressiveness, and the presence of other features of malignancy.
o Can be done outpatient or in physician’s office, or in an operating room.
What is fine-needle aspiration, how is it useful in diagnosing cancer?
o Minimally invasive biopsy, and done by aspirating the target tissue with a needle. It is easy to perform and involves little risk or discomfort to the patient.
o Disadvantages: low volume of cellular components yields false-positive results, and the inability of this method to distinguish b/t invasive and in situ disease.
What is a core biopsy and how is it useful in diagnosing cancer?
o Representative sample of actual target tissue is obtained, with the aid of imaging techniques to ensure that the area of suspicion is correctly targeted and sampled.
o Further definitive surgery may be indicated if sampled tissue is positive for malignancy.
What is sentinel node biopsy and how is it useful in diagnosing cancer?
o New technique advancing the staging and treatment of melanoma, breast, and possibly other malignant tumors.
o NucMed imaging studies are performed that help identify the sentinel node.
o Radioactive material injection, tagged with cancer antigen-specific monoclonal antibody and blue dye into area of tumor. Substance migrates to sentinel node.
o Allows for detection of cancer-affected lymph nodes.
o Sentinel lymph node removal minimizes postop complications such as: pain, numbness, and lymphedema.
o Accurate means of determining stage of breast cancer, w/ fewer complications.
What is axillary node biobsy and how is it useful in diagnosing cancer?
o Critical step in staging breast cancer.
o Surgical procedure typically carried out under general anesthesia.
o Provides needed information at the time of diagnosis about the cancer and its potential for metastasis.
What are the surgical methods for diagnosing and staging cancer
Tissue biopsy, fine needle aspiration, core biopsy, sentinel node biopsy, and axillary node biopsy.
What is radiation therapy? As if we don't already know this......
the use of ionizing radiation in the treatment of patients with benign and malignant diseases. The intent is to deliver a precisely measured dose of radiation to a defined tumor volume with minimal damage to adjacent healthy tissue.
What is radiation oncology?
the medical discipline concerned with the causes, prevention, and treatment of cancer involving special expertise in the therapeutic use of radiation therapy, either alone or in conjunction with surgery, chemotherapy, biotherapy, heat, or oxygen
What is electromagnetic radiation and give examples?
- radiation which consists of associated, interacting electric and magnetic field waves which travel at the speed of light that have no mass but have the appearence of having mass when in tissue. Include: Radio waves, microwaves, visible light, x-rays, and gamma rays.
What is particulate radiation and give examples?
a stream of atomic or subatomic particles that can have a positive charge, a negative charge, or no charge at all, but do have mass. Include: Electrons, protons, neutrons, pi-mesons, and heavy ions.
What is fractionation?
process where radiation is given in smaller doses of a certain period of time
Why is fractionation important in radiation therapy?
Allows the cells to repair between treatments.
What are the 4 R's in radiation therapy?
Repair, redistribution, repopulation, reoxygenation
Mr. Y, a heavy smoker, presents to his family physician with a complaint of a nagging cough continuing for several months. Which of the following examinations is Mr.Y's physician most likely to order?
a. A CT scan of the chest
b. A blood PSA level
c. A chest x-ray
d. An MRI of the chest
C. a chest x-ray
In preparing Mr.Y for tests, patient teaching should include:
a. informatin presented at a level Mr.Y and his family can understand
b. Simple written instructions for preparation
c. Reinforcement of information as needed
d. All of the above
D. all of the above
Mr.Y has completed a series of exams that indicate a lung mass in the right lower lobe. He is scheduled for a CT guided biopsy. Mr Y's daughter asks, "why don't you just go ahead and treat this? You know it's a cancer!" The nurse's most appropriate response would be:
a. we have to be absolutely certain it is cancer before we can treat. The only way to be certain is to have the pathologist look at a sample of the tissue.
b. This institution participates in clinical trials. We are required to document a tissue diagnosis prior to intiation of treatment
c. This test is a part of this work-up. It's not really that big a deal
d. Perhaps you would like to discuss that question with your father's physician.
A. aboslutely certain it is cancer before we treat..blah blah blah
Mrs. S, a patient with breast cancer has complained of back pain and is being evalutated for bone mets. Which of the following test is most likely to be used to evaluate for bone mets?
a. abdominal ultrasound
b.Myelogram
c.Intravenous pyleogram
d.Bone scan
D. bone scan
In planning a community screening program which of the following would be appropriate to include as a screening exam?
a. CEA
b. CA 29.27
c. AFP
d. PSA
D. PSA
This is because PSA is very specific while the others are not.
Grade is a classification system based on :
a. anatomic location of the tumor
b.Size of the original tumor
c.Resectability and location of a malignancy
d.differentiaiton of the cells
D. differentiation of the cells
Staging is nessecary for:
a.comparison of treatment results and research data among facilites
b.planning treatment and prognosis for the induvidual patient
c.comparison of worldwide statistics
d.all of the above
D.all of the above
A reference on a patients chart to a T1 N0 M0 stage malignacy means a the patient:
a. has a small tumor with negative lymphnodes and no mets
b. has a large tumor and status of lymphnodes is unknown
c.has expressed a with that NO one be told about the malignancy
d.has a small tumor with multiple lymphnodes involved and wide spread mets.
A. has a small tumor with negative lymphnodes and no mets
Mrs.Y tells the nurse that she is fearful of the outcome of her breast biopsy. The nurse should:
a. Tell Mrs.Y that ther is nothing to be afriad of; everything will be all right
b. acknowledge the validity of Mrs.Y's feelings and allow her to verbalize them further
c. Tell Mrs.Y since 80% of breast cancer biopsies are negative she has nothing to fear
d.provide Mrs.Y with printed information about breast cancer, the various forms of treatment, and side effects of treatment.
B. acknowledge the validity of Mrs.Y's feelings and allow her to verbalize them further
PET scan is likely to be most useful for tumors that have:
a. a large tumor mass
b. a high metabolic rate
c.a poor blood supply
d. metastisis to the bone
B. A high metabolic rate
What are the various ways that radiation can be delivered to a patient?
External beam, brachytherapy, stereotatic radiosurgery, and radioactive materials through injection or taken orally.
What are radiosensitizers?
agents that enhance radiation damage to tumor cells when they are present at the same time radiation is given. EX: 5-FU, paclitaxel (Taxol), and the nitroimidazoles.
What are radioprotectants?
“radical scavangers” that interact with ionized particles to prevent DNA damage to healthy tissue. Sulfhydryl-containing compounds and Amifostine, which was used to protect normal tissue against radiation and chemo in head, neck, lung, and rectal cancers.
What are the roles of the radiation therapist in patient education?
• Pts need information on what to expect during planning and treatment, the onset and duration of possible side effects, self-care measures, and follow-up care.
What are the general side effects of radiation?
Skin Reactions, fatigue, anorexia, and bone marrow suppression
What is a late effect?
occur months to years after treatment and are generally limited to the area that has been treated. They are often due to damage of blood vessels and connective tissue cells. Many late effects are reduced by fractionating treatment into smaller parts.
What is a side effect?
A problem that occurs when treatment affects healthy tissues or organs, such as: fatigue, pain, nausea, vomiting, decreased blood cell counts, hair loss, and mouth sores. Medical definition: Any unwanted/undesirable nontherapeutic effect caused by a drug or therapy. Peripheral or secondary effects.
What is a complication
A medical problem that occurs during a disease, or after a procedure or treatment. The complication may be caused by the disease, procedure, or treatment or may be unrelated to them. Medical definition-disease or disorder arising as a consequence of another disease.
What is acute?
symptoms or signs that begin and worsen quickly; not chronic.
What is subacute?
a condition between acute and chronic.
What is chronic?
adverse effects on the human body with symptoms that develop slowly over a long period of time or recur.
What are some forman and informal teaching devices that patients can use?
• patients can attend support groups for education, or visit websites.
• For formal education the patient should be provided with approved literature, and easy to understand information and directions from professional medical oncology staff.
The use of radiation therapy to treat cancer is based on the fact that radiation therapy:
a. Is a systemic treatment
b.Is rarely used with chemotherapy
c. Uses heat to alter the cell's DNA
d. Uses high energy radiation
D.Uses high energy radiation
When educating a 70 year-old man about radiation theraoy for prostate cancer, it will be most helpful if he recieves information that:
a. The treatmetn will last approximately 6-7 weeks
b. The actual treatment is short
c. He may be alone in the treatment room for a long time
d.He may feel uncomfortable lying on the treatment table
A. The treatment will last approximately 6-7 weeks
This gentlemen recieving radiation for prostate cancer should also recieve information that during the course of treatment he should:
a.monitor for a cough
b. use a low-residue diet
c. limit fluid intake
d. aviod anticholingeric (WTF?!?) medication
B. Use a low residue diet
When a patien thas recieved a radiopharmacuetical to treat bone mets there:
a. Is not contamintation of the body fluids
b.should be no contact with people in public areas
c. may be temporary increase in blood counts
d. may be a temporary flare in bone pain
D. may be a temporary flare in bone pain
When caring for a hospitalized woman with vaginal implant for cervical cancer:
a. the patient may dangle her legs at the bedside
b.montior placement of the applicator and vaginal packing
c.a nasogastric tube is used for nutrients
d.institute a bowel program to encourage bowem movements
B. monitor placement of applicator and vaginal packing
Recommendations for nursing care of the patient with a radioactive implant includes:
a.use of a lead apron when providing direct patient care
b.conversations with the patient at the bedside
c.use of lead shield at bedside
d.frequent foley catheter care
C. Use of lead shield at bedside
When a moist desquamation of the skin occurs use:
a.Burrow's compress
b. fluorinated steriod cream
c.cornstarch powder
d. mentholated cream
A. Burrow's compress
Recommendations to manage fatigue durign radiation therapy include all except:
a.walking in an excerise program
b.maintaining nutrional status
c.using community resources
d. avoiding naps during the day
D. avoiding naps during the day
When patients experience xerostomia due to salivary gland exposure to radiation therapy, it is recommended that they:
a. use commercial antibacterial mouthwashes
b. use lemon-glycerin swabs
c.preform frequent mouth rinses
d. consume high protein foods such as peanut butter
C. preform frequent mouth rinses
Nausea and vomiting is associated with radiation therapy for:
a. Breast cancer
b. Rectal cancer
c. Pancreatic cancer
d. Brain cancer
C. Pancreatic cancer
What is chemotherapy?
the use of cytotoxic drugs in the treatment of cancer. It is one of the four treatment modalities that provide cure, control, or palliation
What are the 6 ways that chemotherapy can be used?
• Adjuvant therapy-course of chemo used in conjunction with another treatment modality, and aimed at treating micrometastases.

• Neoadjuvant chemotherapy- administration of chemotherapy to shrink tumor before it is removed surgically.


• Primary therapy- treatment of patients who have localized cancer for which an alternative but less than completely effective treatment is available.

• Induction chemotherapy- drug therapy given as the primary treatment for patients who have cancer for which no alternative therapy exists.


• Combination chemotherapy- administration of two or more chemotherapeutic agents to treat cancer; this allows each medication to enhance the action of the other, or to act synergistically with it.

• Myeloblative therapy- dose-intensive therapy used in preparation for peripheral blood stem cell transplantation.
What is the primary focus of chemotherapy?
to prevent cancer cells from multiplying, invading adjacent tissue, or developing metastases.
What are the five phases of the cell cycle?
G1 - post mitotic gap - prepares for DNA synthesis
S - Synthesis - DNA Synthesis actually occurs
G2 - Postsynthesis
M - cell division
G0 = resting phase
What are the principles of chemotherapy in relation to the cell cycle?
• Chemotherapeutic drugs are most active against frequently dividing cells, or in all the phases of the cell cycle, except for G0.
• Chemo is given according to schedules that have proved most effective for tumor kill and that are planned to allow normal cells to recover.
• Normal cells with rapid growth changes that are most commonly affected by chemotherapeutic agents include:
o Bone Marrow (platelets, RBCs, and WBCs)
o Hair Follicles
o Mucosal lining of GI
o Skin
o Germinal cells (sperm and ova)
What is doubling time?
time required for a tumor mass to reach a certain size, or double
What are micrometastisis?
formation of microscopic secondary tumors created by cancerous cells escaping into lymphatic or vascular flow, where they can travel to distant sites.
o b/t 7th and 10th doubling time.
What is the gompertizan function?
pattern of growth that shows that tumors doubling time is more rapid during early growth than at later stages.
What is the pyramid effect?
means by which cancer cells grow. Regulatory mechanism that controls the growth of cancer cells is different from normal cells.
What are factors influencing chemotherapy drug selection and administration?
Blood-Brain Barrier, Chemoprevention, Chronotherapy, cytoprotectants, Liposomes, Radiosensitizers
What is the Blood brain barrier?
Inhibits certain substances entering brain or CNS tumors
o Acts as a screening device, protecting brain and CSF from harmful substances.
o Formed by supporting cells astrocytes and endothelial cells.
o Passage of substances across BBB deoends on:
 Molecular weight, Lipid solubility, Ionizing state
• Large water-soluble charged particles (i.e. glucose)-cannot penetrate BBB
What is chemoprevention?
a pharmacologic intervention that reduces cancer risk in individuals who are highly susceptible to certain cancers by prescribing certain natural or chemical synthetic products or chemotherapy drugs that may reduce or suppress the process of carcinogenesis. Seeks to arrest the process of carcinogenesis before the tumor develops.
What is chronotherapy and what is another name for this?
The practice of administering chemotherapy at certain times of the day that are thought to be optimal for enhanced activity or lessened toxicity. Also called circadian rythm
What is a cytoprotectant?
selectively protect normal tissues from the cytotoxic effects of drugs or irradiationwhile preserving their antitumor effects.
What are liposomes?
Fat molecule made up of layers of lipids, used to convey vaccines, drugs, enzymes, or other substances to target cells or organs. A small spherical particle consisting of a bilayer of phospholipid molecules surrounding an aqueous solution
o Made it possible to manipulate chemotherapy drugs and tailor them to penetrate specific target issues.
What are radiosensitizers?
compunds that enhance the sensitivity of tumors to the effects of radiation, but not to normal tissue.
What are the 2 main classifiactions of chemotherapy drugs?
Cell cycle phase specific and cell cycle phase non-specific
What chemo drugs are cell cycle phase specific and what phase are they specific for?
 Antimetabolites - S phase
 Vinca Alkaloids - M phase
 Misc-asparaginase, dacarbazine
 Antitumor antibiotics (some) varies by drug
• Drugs most effective against actively growing tumors that have a greater proportion of cells cycling through the phase in which the drug attacks the cancer cell
What chemo drugs are cell cycle phase non-specific and give an example?
 Alkalating Agents- Nitrogen Mustard, Cytoxin (Oncovin)
 Antitumor Antibiotics- (some) Bleomycin, Doxirubicin (Adriamycin)
 Nitrosureas- Carmustine (BiCNU)
 Hormones- chemicals secreted by endocrine glands
 Antihormonal agents
 Corticosteroids- androgens, estrogens
What 6 factors are considered for drug selection in chemotherapy?
1. Patient eligibility
2. Cell type
3. Rate of drug absorption
4. Tumor load
5. Tumor location
6. Tumor resistance
What is a cytoprotectant?
selectively protect normal tissues from the cytotoxic effects of drugs or irradiationwhile preserving their antitumor effects.
What is standard dose therapy?
Usual adult dose administered for most cancer patients
What is a cytoprotectant?
selectively protect normal tissues from the cytotoxic effects of drugs or irradiationwhile preserving their antitumor effects.
What are liposomes?
Fat molecule made up of layers of lipids, used to convey vaccines, drugs, enzymes, or other substances to target cells or organs. A small spherical particle consisting of a bilayer of phospholipid molecules surrounding an aqueous solution
o Made it possible to manipulate chemotherapy drugs and tailor them to penetrate specific target issues.
What is a cytoprotectant?
selectively protect normal tissues from the cytotoxic effects of drugs or irradiationwhile preserving their antitumor effects.
What is high dose therapy?
Increased drug dose to achieve tumor cell death
Severe side effects possible
Myelosuppression
What are liposomes?
Fat molecule made up of layers of lipids, used to convey vaccines, drugs, enzymes, or other substances to target cells or organs. A small spherical particle consisting of a bilayer of phospholipid molecules surrounding an aqueous solution
o Made it possible to manipulate chemotherapy drugs and tailor them to penetrate specific target issues.
What are radiosensitizers?
compunds that enhance the sensitivity of tumors to the effects of radiation, but not to normal tissue.
What are liposomes?
Fat molecule made up of layers of lipids, used to convey vaccines, drugs, enzymes, or other substances to target cells or organs. A small spherical particle consisting of a bilayer of phospholipid molecules surrounding an aqueous solution
o Made it possible to manipulate chemotherapy drugs and tailor them to penetrate specific target issues.
What is dose intensity?
doses administered at a higher dose than the standard at a shorter interval
What are radiosensitizers?
compunds that enhance the sensitivity of tumors to the effects of radiation, but not to normal tissue.
What are the 2 main classifiactions of chemotherapy drugs?
Cell cycle phase specific and cell cycle phase non-specific
What are radiosensitizers?
compunds that enhance the sensitivity of tumors to the effects of radiation, but not to normal tissue.
What are the 2 main classifiactions of chemotherapy drugs?
Cell cycle phase specific and cell cycle phase non-specific
What chemo drugs are cell cycle phase specific and what phase are they specific for?
 Antimetabolites - S phase
 Vinca Alkaloids - M phase
 Misc-asparaginase, dacarbazine
 Antitumor antibiotics (some) varies by drug
• Drugs most effective against actively growing tumors that have a greater proportion of cells cycling through the phase in which the drug attacks the cancer cell
What are the 2 main classifiactions of chemotherapy drugs?
Cell cycle phase specific and cell cycle phase non-specific
What chemo drugs are cell cycle phase specific and what phase are they specific for?
 Antimetabolites - S phase
 Vinca Alkaloids - M phase
 Misc-asparaginase, dacarbazine
 Antitumor antibiotics (some) varies by drug
• Drugs most effective against actively growing tumors that have a greater proportion of cells cycling through the phase in which the drug attacks the cancer cell
What chemo drugs are cell cycle phase non-specific and give an example?
 Alkalating Agents- Nitrogen Mustard, Cytoxin (Oncovin)
 Antitumor Antibiotics- (some) Bleomycin, Doxirubicin (Adriamycin)
 Nitrosureas- Carmustine (BiCNU)
 Hormones- chemicals secreted by endocrine glands
 Antihormonal agents
 Corticosteroids- androgens, estrogens
What chemo drugs are cell cycle phase specific and what phase are they specific for?
 Antimetabolites - S phase
 Vinca Alkaloids - M phase
 Misc-asparaginase, dacarbazine
 Antitumor antibiotics (some) varies by drug
• Drugs most effective against actively growing tumors that have a greater proportion of cells cycling through the phase in which the drug attacks the cancer cell
What chemo drugs are cell cycle phase non-specific and give an example?
 Alkalating Agents- Nitrogen Mustard, Cytoxin (Oncovin)
 Antitumor Antibiotics- (some) Bleomycin, Doxirubicin (Adriamycin)
 Nitrosureas- Carmustine (BiCNU)
 Hormones- chemicals secreted by endocrine glands
 Antihormonal agents
 Corticosteroids- androgens, estrogens
What 6 factors are considered for drug selection in chemotherapy?
1. Patient eligibility
2. Cell type
3. Rate of drug absorption
4. Tumor load
5. Tumor location
6. Tumor resistance
What chemo drugs are cell cycle phase non-specific and give an example?
 Alkalating Agents- Nitrogen Mustard, Cytoxin (Oncovin)
 Antitumor Antibiotics- (some) Bleomycin, Doxirubicin (Adriamycin)
 Nitrosureas- Carmustine (BiCNU)
 Hormones- chemicals secreted by endocrine glands
 Antihormonal agents
 Corticosteroids- androgens, estrogens
What 6 factors are considered for drug selection in chemotherapy?
1. Patient eligibility
2. Cell type
3. Rate of drug absorption
4. Tumor load
5. Tumor location
6. Tumor resistance
What is standard dose therapy?
Usual adult dose administered for most cancer patients
What 6 factors are considered for drug selection in chemotherapy?
1. Patient eligibility
2. Cell type
3. Rate of drug absorption
4. Tumor load
5. Tumor location
6. Tumor resistance
What is standard dose therapy?
Usual adult dose administered for most cancer patients
What is standard dose therapy?
Usual adult dose administered for most cancer patients
What is high dose therapy?
Increased drug dose to achieve tumor cell death
Severe side effects possible
Myelosuppression
What is high dose therapy?
Increased drug dose to achieve tumor cell death
Severe side effects possible
Myelosuppression
What is high dose therapy?
Increased drug dose to achieve tumor cell death
Severe side effects possible
Myelosuppression
What is dose intensity?
doses administered at a higher dose than the standard at a shorter interval
What is dose intensity?
doses administered at a higher dose than the standard at a shorter interval
What is dose intensity?
doses administered at a higher dose than the standard at a shorter interval
What is dose density?
o Increased drug doses and combinations of varied drugs
 Doublet or triplet therapy protocols
What are some limitations to chemotherapy agents?
1. Blood brain barrier
2. Drug resistance
3. Injure or kill normal cells while acting on targeted cells
What are 5 reasons to use combination chemotherapy?
1. Maximize cell death
2. Decrease drug resistance
3. Decrease drug toxicity
4. Increase the number of cells killed at various stages or phases of development
5. Increase therapeutic level of drug
What are some general side effects of chemo?
1. Nausea
2. Vomiting
3. Diarrhea
4. Anorexia
5. Hair loss
6. Fatigue
7. Hematopoeitic changes (Anemia, thrombocytopenia)
8. Mucositis
What is a major side effect of doxorubicin(adriamiacin)?
cardiac myopathy
What is a major side effect of bleomycin?
decreased lung compliance
What is a major side effect of cyclophosphomide (cytoxan)?
Hemmoragic cystitis
What are some major side effect of vincristine (oncovin)?
severe constipation and tingling of fingers, toes, and lips
What is a major side effect of cisplatin?
Renal failure
What is nadir?
the period of time where the antineoplastic therapy has its most profound effects on the bone marrow; when the blood counts reach their lowest points.
What are the 6 most common problems with chemotherapy?
• Vesicant Extravasation- accidental leakage of drug into the subcutaneous tissue that causes pain, necrosis, or sloughing of tissue.
• Vesicant
o Agent that can produce a blister, tissue destruction, or both.
• Irritant
o Agent that can cause aching, tighness, and phlebitisat the injection site or along the vein line w/ or w/o an inflammatory reaction.
• Flare
o Local allergic reaction w/o pain that is usually accompanied by red blotched along the vein line.
• Delayed extravasation
o One in which symptoms occur 48 hours or more after the drug is administered.
• Tissue necrosis
o Pain or burning at the IV site, progressing to erythema, edema, and superficial skin loss.
• Anaphylaxis
o A severe, whole-body allergic reaction to a chemical that has become an allergen
o Anxiety, hypotension, cyanosis, respiratory distress, abdominal cramping, flush, chills and urticaria.
What is biotherapy?
treatment with agents derived from biological sources or affecting biological response. Many derived from mammalian genome
What is Immunotherapy?
the prevention or treatment of disease with substances that stimulate the immune response
What is active specific immunotherapy?
immunization with tumor cells or tumor cell extracts, such as proteins or gangliosides administered as tumor antigens, or vaccines.
What is active non specific immunotherapy?
an attempt to boost overall immunity through the use of adjuvants such as bacterial extracts.
What is passive immunotherapy?
the administration or transfer of previously sensitized immunologic reagents such as antisera (which contain sensitized antibodies) or immune-reactive cells to a tumor-bearing host.
What is adoptive immunotherapy?
the passive transfer of sensitized cells such as lymphocytes or macrophages.
Describe interferons
- family of glycoprotein hormones possessing pleiotrophic biologic effects. All IFNs mediate their cellular effects after binding to a specific receptor.
o Wide range of biologica effects including: antiviral, antiproliferative, and immunomodulatory.
o Antiviral effects- renders uninfected cells resistant to attack by offending virus as well as by a variety of other viruses. Internalization of the IFN-receptor complex causes a sequence of events that produces antiviral proteins and enzymes.
o Alpha
 Antiviral, antiproliferative, and immunomodulatory effects
 Inhibits cellular replication of viral DNA, enhances natural killer cell activity
 B cells, T cells, macrophages, null cells in response to viruses or tumor cells
 Hairy cell leukemia, AIDS related Kaposi’s sarcoma and many other cancers

o Beta
o Gamma
Describe lymphokines
• Lymphokines-Interleukins-IL-2 is a glycoprotein produced by activated T-helper cells and is a potent modulator of immune response. Can reverse immune deficiency.
o IL-2 exerts its biologic effects by binding to membrane-bound receptors on certain immune cells. IL-2 supports the growth and maturation of subpopulations of T cells both in vitro and in vivo, stimulates cytotoxic T cells, stimulates the proliferation and activity of NK cells, and develops the capacity in lymphoid cells incubated with IL-2 to l yse fresh tumor cells. These cells are known as lymphokine-activated killer (LAK) cells.
o First response action: activation of T-cells by antigen or mitogen.
o Second response: interaction with IL-1.

o Neumega (oprelvekin) - platelet growth factor
 Stimulates megakaryocye and thrombocyte production
 Reduces the number of platelet transfusions needed after chemotherapy
 SubQ 21 days
o The group of proteins in the body that produce the WBCs
o IL-2 – 1976
o Sub Q or IV frequently
Describe hematopoetic growth factors
• Hematopoietic Growth Factors- family of glycoproteins responsible for proliferation, differentiation, and maturation of hematopoietic cells in vitro. They also stimulate fx of certain mature leukocytes. “Colony Stimulating Factors”
o Stimulate or regulate the growth, maturation, and differentiation of bone marrow stem cells
 Decreases the rebound time of bone marrow products after chemotherapy or marrow transplant
 Enhances immune system to fight tumors
 Permits higher dose of drugs
What are monoclonal antibodies?
individual B cells produce an antibody specific for single antigenic determinants; therefore when an antigen invade the body, a variety of antibodies against it are produced MoAb.
o Mediate an antitumor cytotoxic effect through complement-dependent cytotoxicity, or antibody-dependent cellular toxicity.
o Use MoAb directed against cell surface receptors involved in proliferation, such as epidermal growth factor receptor (EGFR).
o Trastuzumab (Herceptin)
o Breast cancers resistant or refractory to use of paclitaxel (Taxol)
o IV, weekly
What is radioimmunotherapy?
combines radioactive isotopes such as Iodine-131 (I-131) and yttrium-90 (Y-90) with a MoAb. The radioisotope is carried to the tumor by the MoAb that attaches to a specific antigen present on the tumor cell surface. Once at the tumor sight, radiation is targeted to tumor with the surrounding normal cells receiving less radiation that in they were exposed to EBRT. Cancer cells are destroyed by the combination of targeted radiation therapy, the biologic effects of the MoAb, and the cross fire effect of the radiation on nearby tumor cells to which the antibody didn’t bind.
What are angiogenesis inhibitors?
drugs that inhibit the formation of blood vessels, thus cutting off the supply of nutrients and oxygen to malignant tumors, resulting in their death.
What are Epidermal growth factor receptor/tyrosine kinase inhibitors?
responsible for activating multiple downstream signaling pathways governing tumor growth.
What are Stem cells?
An undifferentiated cell of a multicellular organism that is capable of giving rise to indefinitely more cells of the same type, and from which certain other kinds of cell arise by differentiation
What is an autologus transplant?
a transplant in which the the patient’s own bone marrow or stem cells are collected (harvested), placed in frozen storage (cytopreserved), and reinfused into the patient after the conditioning regimen
What is a syngenetic transplant?
- the donor is the patients identical twin, having identical matched cell type.
What is an allogeneic transplant?
a transplant in which the patient receives someone else’s bone marrow or stem cells. 3 types: syngenic, related, and unrelated.
What is apherisis?
using standard, commercially available cell separators, that are programmed to collect either lymphocytes or low density leukocytes. The remaining blood components are returned to the patient. Peripheral blood stem cells (PBSCs) are collected by this process.
What is mobilization?
the use of chemo and growth factors together in order to stimulate stem cells, creating an increase in the number of stem cells in the blood and lengthening the time they are present.
What is bone marrow transplantation?
Bone marrow transplant is the IV infusion of hematopoetic stem cells (progenitor cells) to re-establish marrow function and reset immune function.
What is a stem cell transplantaion?
the process of replacing diseased or damaged bone marrow with normally functioning bone marrow.
What are 3 sources of stem cells?
• Bone marrow harvest procedure
o General anesthetic
o Outpatient surgery
o Multiple bone marrow aspirations
• Peripheral blood
o Daily injections of G-CSF
o Large bore IV access
o Pheresis
• Cord Blood
o Placental blood at birth
What are the most common malignant diseases treated with stem cells?
• Acute myelogenous leukemia
• Non-Hodgkin’s lymphoma
• Acute lymphocytic leukemia
• Juvenile myelomonocytic leukemia
• Chronic myelogenous leukemia
• Myelodysplastic syndrome
• Hodgkin’s disease
• Renal cell carcinoma
• Neuroblastoma
What are the most common non malignant diseases treated with stem cells?
• aplastic anemia
• myelofibrosis
• wiskott-Aldrich syndrome
• SCIDS
• Mucopolysacharoidosis
• Osteopetrosis
• Lipid storage diseases
What is the hemepoitic stem cell process?
• Pretreatment work-up
• Conditioning regimens
• Nonmyeloablative conditioning regimens
• Transplantation of marrow, stem cells, and cord blood
• Engraftment period
What are the complications of the hemeopoetic stem cell process?
• Infection
• Pulmonary complications
• Veno-occlusive disease
• Graft-versus-host disease
• Chronic GVHD
o New immune system recognizes host tissue as “non-self”
 Acute-GI, Liver, Skin, lung, hematologic most common
• Treatment with steroids, immunosuppressant
 Chronic – GI, liver, skin, lung, ocular,
• Treatment steroids, immunosuppressant, puva therapy, bcell directed therapy (rituximab)
What is disseminated intravascular coagulation?
• Stimulated by tumor or infection
• Signs & Symptoms
▫ Frank bleeding, bruising, petechiae, guiac-positive stool or emesis, hemoptysis
• Treatment
▫ Blood and blood products
 RBCs, platelets, fresh frozen plasma or cryoprecipitate