Biological Therapies



Biological Therapies


Brenda K. Shelton



I. History

A. In 1895, Hericourt and Richet were first to document antitumor immune therapy in humans, defining the immune surveillance theory of cancer development.

B. Primary barrier to development of biological therapy as a cancer treatment modality was ability to generate large quantities of immune substances, resolved by development of recombinant DNA technology in the 1970s.


II. Definitions/Overview

A. Immune activity is the essential homeostatic mechanism protecting a person against potential pathogens and foreign tissues.

B. Biological therapy is defined as use of naturally occurring immune substances to affect the body’s immune responses. Includes activities such as:

1. Immune stimulation

2. Immune augmentation

3. Immune suppression


III. Rationale for Use

A. Proposed Physiologic Activity

1. The discovery of individual tumor-surface antigens (tumor-specific antigens) and tumor-associated antigens has supported the belief that most human cancers have antigens capable of eliciting an immune response.

2. Cells may spontaneously transform to malignancy or mutate after exposure to carcinogens.

3. Once malignant transformation has occurred, the immunologic surveillance system recognizes the neoplastic cell as foreign by the presence of antigens on the surface of the tumor cells.

4. Under normal circumstances, the immune surveillance system destroys the malignant cell in the preclinical stage; however, an impaired immune system will permit malignant cells to proliferate to clinical disease.

B. Evidence Supporting Immunosurveillance Theory

1. Higher incidence rates of cancer in children, elderly, people with immunodeficiency diseases, and in organ transplantation patients.

2. Reports of spontaneous tumor regression.

3. It has also been shown that tumors heavily infiltrated by lymphocytes may have a better prognosis than those that do not demonstrate this characteristic (Reiger, 2001).


C. Antitumor biological agents have been developed based on different proposed mechanisms of altering the host-tumor response so tumor cells are immunologically destroyed.


IV. Biology of Therapy

A. Basic Goals of Anticancer Biotherapy

1. To stimulate direct and indirect immunocompetence

2. To create tumor-specific immunity

3. To induce tumor regression when used adjunctively with other cancer treatments

B. Indications for Biological Therapy

1. Known tumor-associated antigen that can be targeted for destruction (eg, her-2-neu receptor in breast cancer patients).

2. Tumors with high lymphocytic infiltration.

3. Limited disease thought to be more responsive to immunologic manipulation.

4. Tumors for which spontaneous remissions have been reported (eg, malignant melanoma, renal cell carcinoma).

5. Existing or anticipated bone marrow suppression is the clear single indication for restorative biological therapy.

6. Prevention of rejection in organ transplant recipients and suppression of autoimmune disease mechanisms are indications for immunosuppressive immune modulators or monoclonals that target the T lymphocytes.

C. Principles to Determine Patient Eligibility

1. Tumor size: Large tumors are thought to depress the immune system, and less likely to demonstrate an antitumor response with biological therapy because of the slow immunologic destruction induced by these agents.

2. Tumor type: Patients who derive the most benefit from biological therapy have disease that has not progressed beyond stage II and who are not severely immunosuppressed.

3. Immunocompetence: Patients able to generate positive skin test reactions demonstrating active immune responses may have better antitumor response rates. Quantitative assessment of immune function is made by peripheral blood counts, immunoglobulin assays, and bone marrow aspiration.

D. Biological Therapy Administration

1. Treatment scheduling: Treatment should be timed carefully with other anticancer therapies, so the immune system can recover from the effects of these other modalities. Certain chemotherapeutic agents demonstrate synergistic antitumor activity with biotherapeutic agents (eg, 5-fluorouracil and levamisole).

2. Dosage

a. Biotherapy is not dosed according to maximal tolerated dose (MTD).

b. Biological therapy may exert its optimal biological effect at a dose much lower than the MTD.

c. Optimal biological dose (OBD)—the dose which, with a minimum of side effects, produces the optimal desired clinical responses.

3. Evaluation of efficacy


a. Traditional measures of antineoplastic therapy measure tumor size as the primary assessment of successful therapy.

b. Biotherapeutic trials must incorporate measures of biological effects on the immune system, and immunologic assays.

c. Stabilization of disease may be a more accurate objective than reduction of tumor because the objective of biological therapy is immune activation.


V. Classification of Biological Agents:

There is no defined classification system of biological agents because many have multiple immunologic antitumor activities. The general categories and their nursing implications are listed in Table 4-1.

A. Antitumor biotherapy uses active and passive mechanisms to enhance the nonspecific and specific host-tumor immune responses through the use of attenuated live bacteria or their products or through immunochemical agents. Antitumor biotherapy is further subdivided into cytokines, vaccines, and monoclonal antibodies.

1. Cytokines are cell-killer substances modeled after normal human lymphocyte secretions that are administered to enhance normal immune defense mechanisms. Human growth factors (eg, erythropoietin, granulocyte colony stimulating factor, granulocyte-macrophage colony stimulating factor, and platelet colony stimulating factor) are termed cytokines in some texts, but they do not have antitumor properties and are, therefore, described as restorative biotherapy here.

a. Interferons—alfa, beta, gamma

b. Interleukins—interleukin-2

c. Other—tumor necrosis factors (still investigational)

2. Antitumor vaccines are antibodies specifically engineered to destroy particular tumor antigens, theoretically making them tumor-specific, long-lived, and nontoxic. A major problem in vaccine therapy is the identification and purification of relevant antigens to be used in preparing vaccines.

a. Vaccines may be made from animal or human models.

b. Types of cancer vaccines determined by the transport mechanism of the vaccine: recombinant bacteria, protein peptides, nucleic acids, recombinant viruses, transduced cells, modified tumor cells (Kinzler & Brown, 2001).

c. Immunoadjuvants involve the use of transports that primarily activate existing host immune activities rather than replace defective gene sequences (eg, bacteria, interleukin-12, granulocyte-macrophage colony stimulating factor). This category includes the single vaccine product currently licensed as an anticancer therapy (bacillus Calmette-Guérin [BCG]).

d. Dendritic cells are potent antigen-presenting cells that are essential for recognition and destruction of tumor cells. Removal and cytokine activation of human dendritic cells followed by reinfusion is thought to produce effects similar to other immunoadjuvant therapies (Kinzler & Brown, 2001).

3. Monoclonal antibodies are antibodies derived from the fusion of an antibody-producing cell, such as a B lymphocyte, and another cell.














TABLE 4-1 Biotherapy Agents*

























































Biologic Classification and Agents


Adverse Effects


Special Nursing Implications


Antitumor biotherapy: vaccine




  • Bacillus Calmette-Guérin


Antitumor biotherapy: cytokines




  • Interferon-alfa



  • Interferon-beta



  • Interferon-gamma



  • Interleukin-2 (IL-2)




  • Flulike syndrome within 12-24 hours after dose; subsides within 48 hours




    • Injection site irritation, wheal reaction



    • Dysuria, urinary urgency or frequency if administered via the bladder



    • Flulike syndrome: fever, chills, myalgias, arthralgias, headache



    • Gastrointestinal distress: anorexia, nausea, vomiting, diarrhea



    • Elevated liver enzymes



    • Weight loss



    • Primarily IL-2


      – Capillary leak syndrome (edema, vascular volume depletion, hypotension)


      – Oliguria and renal insufficiency


      – Dysrhythmias


      – Flushing, erythema, rash, pruritus


      – Thrombocytopenia


      – Cognitive dysfunction: difficulty concentrating, emotional lability, confusion, hallucinations




  • Protect from light.



  • Do not use alcohol to prepare skin because it can kill organisms.




    • Avoid concomitant administration of isoniazid because it may inhibit BCG multiplication.



    • Teach patient self-injection technique if applicable.



    • Reconstituted, most agents stable up to 1 month in refrigerator.



    • Prevent flu symptoms with prophylactic acetaminophen or nonsteroidal antiinflammatory agents.



    • Pretreat injection site with ice or massage to reduce injection site irritation.



    • IL-2


      – Perform baseline cardiac and pulmonary function tests before therapy.


      – Give cautiously in neurologic diseases (eg, seizure disorder), brain metastases.


      – Be aware of wide variation in dose range, schedule, and mode of administration, affecting variability of adverse effects.


      – Adding albumin to aldesleukin injectable is also thought to reduce injection site irritation.


      – Use a large variety of symptom management strategies (eg, guided imagery) and concomitant medications (eg, diuretics, antipyretics, antiemetics) to abrogate adverse effects and continue therapy.



    • Adverse effects are completely reversed with cessation of therapy.


Cytomodulatory biologic therapy; monoclonal antibodies for diagnosis




  • Satumomab pendetide (OncoScint CR/OV)



  • Carpromab pendetide (ProstaScint)




  • Fever, chills



  • Hypersensitivity (rare)




  • Detection of ovarian or colorectal cancer (OncoScint), or prostate cancer (ProstaScint)



  • Premedicate with acetaminophen for flulike symptoms



  • Have emergency equipment available for rare circumstance of anaphylaxis.


Antitumor biologic therapy; monoclonal antibodies




  • Alemtuzumab (Campath)



  • Gemtuzumab (Mylotarg) [combined with antitumor antibiotic]




    • Rituximab (Rituxan)



    • Trastuzumab (Herceptin)




  • Infusion-related chills or fever, hypotension



  • Hypersensitivity reaction (pruritus, rash, dyspnea, wheezing)



  • Nausea, vomiting



  • Pain at disease sites



  • Fatigue



  • Elevated liver enzymes



  • Bone marrow suppression—neutropenia, thrombocytopenia




  • Administer as IV infusion only.



  • Stable for 24 hours after reconstitution if remains unrefrigerated.



  • Premedication with acetaminophen and diphenhydramine is recommended.



  • Vital signs q 15 min 3 1 hr, then q 30 min during infusion.



  • Have emergency equipment readily available during infusion in case severe hypersensitivity occurs.



  • Trastuzumab has been associated with acute onset of car diomyopathy and congestive heart failure, especially when administered with anthracyclines or cyclophosphamide


Cytomodulatory biologic therapy; radio-labeled monoclonal antibodies




  • Tositumomab + Iodine131 (Bexxar)



  • 90Y ibritumomab tiuxetan (Zevalin)



  • Nofetumomab + technetium (Verluma)




  • Acute hypersensitivity reactions—fever, chills, hypotension, bronchospasm



  • Bone marrow suppression—neutropenia, thrombocytopenia



  • Asthenia (Zevalin only)




  • Must be administered by radiation therapy professionals



  • Observe the patient continuously for the first 15 minutes.



  • Monitor vital signs every 15 minutes for at least an hour.



  • Have emergency equipment available for rare circumstance of anaphylaxis.



  • Monitor for infection or bleeding.


Cytomodulatory biologic therapy; T-lymphocyte/TNF suppressing monoclonal antibodies




  • Adalimumab (Humira)



  • Daclizumab (Zenapax)



  • Infliximab (Remicade) [anti-TNF antibody]



  • Muromonab-CD3 (Orthoclone OKT3)



  • Natalizumab (Antegren)



  • Omalizumab (Volair) [allergen antibodies]




  • Infection




  • Implement infection prevention precautions.



  • Discontinue other antirejection drugs to reduce the risk of infection.


Cytomodulatory biologic therapy; fusion proteins




  • Denileukin difitox (Ontak) [diphtheria toxin and interleukin-2]




  • Acute hypersensitivity reactions—fever, chills, hypotension, bronchospasm



  • Capillary leak syndrome (edema, vascular volume depletion, hypotension)



  • Hypoalbuminemia about 1 week after administration




  • Observe the patient continuously for the first 15 minutes.



  • Monitor vital signs every 15 minutes for at least an hour.



  • Have emergency equipment available for rare circumstance of anaphylaxis.



  • Plan for monitoring blood pressure, weight, and presence of edema.


Cytomodulatory biologic therapy; polypeptide antibiotics




  • Cyclosporine



  • Tacrolimus




  • Hypertension



  • Mental status changes



  • Decreased seizure threshold.



  • Nausea, vomiting



  • Renal dysfunction




  • Avoid other drugs with neurotoxicity.



  • Monitor blood levels as ordered.



  • Monitor BUN and creatinine during therapy, and report elevations in laboratory values that may necessitate dose reduction.



  • Reduce dose slowly while observing for signs and symptoms of organ rejection.



  • Monitor BP frequently.



  • Administer oral cyclosporine with chocolate milk to mask taste and maximize absorption.


Restorative biologic therapy; hematopoietic growth factors




  • Erythropoietin



  • Granulocyte colony stimulating factor (G-CSF)



  • Oprelvekin (interleukin-11)



  • Granulocyte-macrophage colony stimulating factor (GM-CSF)




  • RBC growth factors: headache, hypertension, increased clotting tendency



  • WBC growth factors: fever, chills, bone pain, leukocytosis



  • Platelet growth factor: atrial arrhythmias Blurred vision, dyspnea Fluid retention (eg, edema, increased effusions)



  • All: injection site erythema or irritation




  • Indications for usage are lineage specific.



  • Erythropoietin:


    – Monitor reticulocyte count for drug effectiveness, and reduce dose for dramatic increases in RBC count.


    – Monitor and replenish iron or transferrin for greater efficacy of treatment.


    – Monitor BP for hypertension.


    – Monitor for thromboses or risk factors for clotting



  • WBC growth factors:


    – Administer 24 hours after conclusion of antineoplastic therapy.


    – Use acetaminophen or nonsteroidal antiinflammatory agents prophylactically to reduce risk or severity of flu symptoms.


Restorative biologic therapy; hematopoietic growth factors





  • Platelet growth factors:


    – Administer 6-24 hours after conclusion of antineoplastic therapy and continue after the nadir of the platelet count.


    – Monitor platelet count daily during therapy



  • All:


    – Pretreat with ice or massage injection site to reduce irritation.


Cytomodulatory biologic therapy; retinoids




  • Bexarotene (Targretin)



  • Alitretinoin (Panretin, 9-cis-retinoic acid)



  • Tretinoin (Vesanoid, All-trans-retinoic acid, ATRA)



  • Tretinoin liposomal




  • Erythema, skin irritation with topical administration



  • Vitamin A toxicity—headache, fever, dry skin and mucous membranes, pruritus, bone pain, nausea and vomiting, visual disturbances



  • Systemic administration of ATRA produces “ATRA syndrome”—fever, dyspnea, weight gain, pulmonary infiltrates, effusions, hypotension



  • Systemic administration may cause:


    – Temporary (rarely permanent) hearing loss


    – Alopecia


    – Agitation, emotional disturbances, forgetfulness, paresthesias


    – Increased cholesterol or triglycerides




  • Assess baseline skin integrity before applying topical agents.



  • Use gloves to administer any of these agents topically.



  • Check for hypersensitivity to any previous retinoids (eg, acne treatments, vitamin A supplements)



  • When giving systemic retinoids, check hepatic enzymes.



  • Observe for “ATRA syndrome” and treat symptomatically while discontinuing the drug.


Cytomodulatory biologic therapy; anti-angiogenesis factors




  • Thalidomide (Thalomid)




  • Flulike syndrome—fever, headache, arthralgias, myalgias



  • Neuropathy



  • Constipation




  • Monitor complete blood count for drug toxicity



  • Monitor for skin reactions and treat rashes symptomatically.



  • Provide patient education about risk to the fetus if this drug is consumed while pregnant.


Cytomodulatory biologic therapy; miscellaneous




  • Levamisole




  • Flulike syndrome—fever, headache, arthralgias, myalgias



  • Fatigue



  • Gastrointestinal distress: stomatitis, altered taste sensations, nausea, vomiting, abdominal pain, diarrhea



  • Bone marrow suppression: neutropenia, thrombocytopenia



  • Dizziness




  • Always give exactly on time to enhance synergistic properties with 5-fluorouracil.



  • Concomitant alcohol consumption can cause disulfiram reactions.



  • Inhibits cholinergic activity.



  • Monitor phenytoin levels; may be higher while receiving levamisole.


*Specific clinical indications for each agent are included in disease-specific chapters, and this table describes an overview of the general clinical features of each category of agents.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Sep 16, 2016 | Posted by in NURSING | Comments Off on Biological Therapies

Full access? Get Clinical Tree

Get Clinical Tree app for offline access