Expired activity
Please go to the PowerPak homepage and select a course.

Pharmacotherapy Review 2021: Oncologic Disorders

INTRODUCTION

The year 2020 was unprecedented year in many ways, including the number and importance of advances in oncology. Even as the world focused on a coronavirus pandemic, more than 20 new drugs with oncology indications were approved by the U.S Food and Drug Administration (FDA) (Table 1).1 In addition, 30 new indications were approved for existing oncology drugs, most of them for immunotherapy drugs, specifically checkpoint inhibitors. Additionally, advancements in most types of hematologic cancers and solid tumors have occurred as well as cancer prevention, screening, and supportive care.

In this continuing education program for generalist pharmacists, key oncologic advancements of 2020 are detailed.

Table 1. 2020 New Drug Approvals in Oncology1
Generic Names (Brand Names) Drug Classes Indications Approval Dates
Avapritinib (Ayvakit) Oral tyrosine kinase inhibitor of PDGFRA, PDGFRA D842 mutations and multiple KIT exon 11, 11/17 and 17 mutants Adults with unresectable or metastatic GIST harboring a PDGFRA exon 18 mutation, including D842V mutations 1/9/20
Azacitidine (Onureg) Oral pyrimidine nucleoside cytidine analog (cytotoxic agent) Maintenance treatment of AML patients achieving complete remission (± blood count recovery) following induction chemotherapy 9/1/20
Belantamab mafodotin-blmf (Blenrep) Antibody-drug conjugate; antibody directed against BCMA found on normal B lymphocytes and myeloma cells and microtubular inhibitor (cytotoxic agent) MMAF Adults with relapsed or refractory multiple myeloma who have received 4 prior therapies, including an anti-CD38 monoclonal antibody, proteasome inhibitor, and an immunomodulatory drug 8/5/20
Brexucabtagene autoleucel (Tecartus) CD19-directed genetically modified autologous T cell immunotherapy Adults with relapsed or refractory mantle cell lymphoma 7/24/20
Capmatinib (Tabrecta) Oral tyrosine kinase inhibitor of MET Adults with metastatic NSCLC whose tumors have a mutation leading to MET exon 14 skipping 5/6/20
Daratumumab and hyaluronidase-fihj (Darzalex Faspro) CD38 monoclonal antibody Adults with newly diagnosed or relapsed or refractory multiple myeloma 5/1/20
Decitabine/ cedazuridine (Inqovi) Oral nucleoside metabolic inhibitor (decitabine) combined with cytidine deaminase inhibitor (cedazuridine) (cytotoxic agent) Adults with MDS 7/7/20
Isatuximab-irfc (Sarclisa) IgG1-derived monoclonal antibody directed at CD38 Adults with multiple myeloma who have received >2 therapies including lenalidomide and a proteasome inhibitor in combination with pomalidomide and dexamethasone 3/2/20
Lurbinectedin (Zepzelca) Alkylating agent (cytotoxic agent) Adults with metastatic small-cell lung cancer after disease progression with platinum-based therapy 6/15/20
Luspatercept-aamt (Reblozyl) Recombinant fusion protein binds endogenous tumor growth factor beta, diminishing Smad 2/3 signaling and increasing erythroid maturation Treatment of anemia failing an erythropoiesis-stimulating agent or 2 or more packed red blood cells over 8 weeks in adults with very low- to intermediate-risk MDS with ring sideroblasts, or MDS/myeloproliferative neoplasm with ring sideroblasts and thrombocytosis 4/3/20
Margetuximab-cmkb (Margenza) HER2/neu receptor antagonist, chimeric Fc-engineered IgG1 kappa monoclonal antibody Adults with metastatic HER2-positive breast cancer who have received 2 or more prior anti-HER2 regimens, at least 1 of which for metastatic disease in combination with chemotherapy 12/16/20
Mitomycin gel (Jelmyto) DNA synthesis inhibitor (cytotoxic agent) Adults with low-grade upper tract urothelial cancer 4/15/20
Naxitamab-gqgk (Danyelza) GD2-binding monoclonal antibody Adult and pediatric patients 1 year of age and older with relapsed or refractory high-risk neuroblastoma in the bone or bone marrow demonstrating a partial response, minor response, or stable disease prior to therapy in combination with GM-CSF 11/25/20
Pemigatinib (Pemazyre) Oral tyrosine kinase inhibitor of FGFR 1, 2, and 3 Adults with previously treated, unresectable locally advanced or metastatic cholangiocarcinoma with FGFR2 fusion or other rearrangement 4/20/20
Pertuzumab, trastuzumab, hyaluronidase-zzxf (Phesgo) Fixed-dose subcutaneous combination of HER2 monoclonal antibodies Neoadjuvant treatment of HER2-positive locally advanced, inflammatory or early-stage breast cancer and adjuvant treatment of HER2-positive early breast cancer at high risk of recurrence 6/29/20
Pralsetinib (Gavreto) Oral tyrosine kinase inhibitor of wild-type RET and oncogenic RET fusions and mutations Adults with metastatic RET fusion-positive NSCLC; patients older than 12 years with advanced or metastatic RET-mutant medullary thyroid cancer who require systemic therapy; patients older than 12 years with advanced or metastatic RET fusion-positive thyroid cancer who require systemic therapy and are radioactive iodine-refractory (if radioactive iodine is appropriate) 9/4/20
Relugolix (Orgovyx) Nonpeptide GnRH receptor antagonist that competitively binds to pituitary GnRH receptors Adult patients with advanced prostate cancer 12/18/20
Ripretinib (Qinlock) Oral tyrosine kinase inhibitor of KIT proto-oncogene and PDGFRA kinase Adults with advanced GIST who have progressed after 3 or more treatments 5/15/20
Sactuzumab govitecan-hziy (Trodelvy) Trop-2 directed antibody-drug conjugate; monoclonal antibody recognizes Trop-2 (sactuzumab) and topoisomerase I inhibitor, SN-38 (cytotoxic agent) Adults with triple-negative breast cancer who have received 2 or more therapies for metastatic disease 4/22/20
Selpercatinib (Retevmo) Oral tyrosine kinase inhibitor of wild-type RET, mutated RET, and VEGFR1 and 3 Adults with RET fusion-positive NSCLC, adult and pediatric patients (>12 yr) with advanced or metastatic RET-mutant medullary thyroid cancer; adult and pediatric patients (>12 yr) with advanced or metastatic RET fusion-positive thyroid cancer who require systemic therapy and are radioactive iodine-refractory 5/8/20
Selumetinib (Koselugo) Oral tyrosine kinase inhibitor of MEK 1 and 2 Pediatrics (>2 yr) with neurofibromatosis type 1 who have symptomatic, inoperable plexiform neurofibromas 4/10/20
Tafasitamab-cxix (Monjuvi) CD19-directed cytolytic monoclonal antibody Adults with relapsed or refractory diffuse large B-cell lymphoma not eligible for autologous HSCT in combination with lenalidomide 7/31/20
Tazemetostat (Tazverik) Oral tyrosine kinase inhibitor of methyltransferase, EZH2, and some EHZ2 gain-of-function mutations Adults and pediatrics (>16 yr) with metastatic or locally advanced epithelioid sarcoma not eligible for complete resection; adults with relapsed or refractory follicular lymphoma whose tumors are positive for EZH2 mutation and have received at least 2 prior systemic therapies; adult patients with relapsed or refractory follicular lymphoma who have no satisfactory alternative treatment options 1/23/20
Tucatinib (Tukysa) Oral tyrosine kinase inhibitor of HER2 Adults with advanced or unresectable or metastatic HER-2 positive breast cancer, including those with brain metastases who have received >1 anti-HER2-based regimens in metastatic setting in combination with trastuzumab and capecitabine 4/17/20
Abbreviations used: AML, acute myeloid leukemia; CD, cluster of differentiation; FGFR, fibroblast growth factor receptor; FOLFIRI, folinic acid (leucovorin), fluorouracil, irinotecan hydrochloride; GIST, gastrointestinal stromal tumor; GM-CSF, granulocyte macrophage colony stimulating factor; GnRH, gonadotropin releasing hormone; HSCT, hematopoietic stem cell transplantation; IgG1, immunoglobulin g G1; MEK, mitogen-activated protein kinase; MET, mesenchymal-epithelial transition; MDS, myelodysplastic syndrome; NSCLC, non-small cell lung cancer; PDGFRA, platelet-derived growth factor receptor A; PD-L1, programmed death 1 ligand; VEGFR, vascular endothelial growth factor receptor.

Immunotherapy Update

Normally, the immune system’s role is to recognize and destroy foreign cells, including cancer cells.2 However, cancer cells have created maneuvers to trick the immune system and avoid being destroyed, commonly known as mechanisms of resistance. Some examples of resistance mechanisms include: 1) genetic changes that allow cancer cells to hide from the immune system 2) the addition of surface proteins, allowing for downregulating the activity of immune cells, and 3) change in the healthy cells surrounding the tumor, which alters the immune system’s activity against the cancer cells. To better combat these setbacks, immunotherapy can be used to aid the immune system in killing cancer cells.

The era of cancer immunotherapy is not new. In fact, it began nearly 125 years ago when William Coley discovered the induction of inflammation by direct tumor inoculation of bacterial products.3 It was not until the 1980s, however, that the first immunotherapy, interleukin-2, was approved by FDA. Although interleukin-2 was highly effective in some patients with cancer, its widespread use was limited by its severe toxicity. Other immunotherapies such as interferons also had limited use due to efficacy and safety.

The second immunotherapy breakthrough occurred nearly 2 decades later, when checkpoint blockade immunotherapy was discovered. The initial finding was that cytotoxic T-lymphocyte associated protein 4 (CTLA-4) blocking antibodies enhanced antitumor immunity.3 Another important checkpoint of the immune system is the programmed cell death receptor 1 (PD-1) molecule, which negatively regulates T cell effector function following binding to its ligand, PD-L1. PD-L1 is expressed on the surface of tumor cells or other immune cells.

Both PD-1 and PD-L1 antibodies are on the market and widely used to treat many cancers. Ipilimumab (Yervoy) is the sole drug currently in the CTLA-4 inhibitor drug class. PD-1 inhibitor drugs currently include nivolumab (Opdivo), pembrolizumab (Keytruda) and cemiplimab-rwlc (Libtayo). PD-L1 inhibitors include atezolizumab (Tecentriq), avelumab (Bavencio), and durvalumab (Imfinzi).

As of 2020, these medications are no longer considered “last ditch” agents, as they have been in the past. These therapies are now often used as first- or second-line therapy in some advanced forms of cancer and can also be considered as adjuvant therapy in some early-stage cancers. Additionally, checkpoint inhibitors are now indicated in many different types of solid tumors and hematologic malignancies as monotherapy or as combination therapy (see Tables 2, 3, and 4).4-10

Initially, checkpoint inhibitors were evaluated and approved as monotherapy. However, as the understanding of the complex immunosuppressive nature of cancer unfolds, novel combination strategies targeting the various elements have been found to be successful in many solid tumors (see Table 4).

Similarly, immunotherapy is being used as maintenance therapy in some cancers after a response has been achieved with chemotherapy. Examples are avelumab for locally advanced or metastatic urothelial carcinoma following first-line platinum-based therapy and durvalumab for stage III non-small cell lung cancer patients following adjuvant concurrent platinum-based chemotherapy and radiation therapy.9,10 Interest is growing in the use of immune checkpoint inhibitor maintenance therapy as a strategy for prolonging the benefits of first-line therapy while minimizing toxicity.

Another advancement in checkpoint inhibitor therapy is the extended-dosing interval (see Table 5). Initially, checkpoint inhibitors were approved in dosing intervals of every 2 or 3 weeks. As of 2020, atezolizumab, nivolumab, pembrolizumab, ipilimumab, and durvalumab have been approved for extended dosing intervals.4-7,9 Evidence supporting extended-interval dosing regimens is still preliminary, but this could minimize the risk of hospital-acquired transmission of infections, minimize work burden for oncology nursing/staff, and improve patient quality of life.11 These extended-dosing intervals were approved under accelerated approval based on tumor response rate, durability of response, pharmacokinetic data, the relationship of exposure to efficacy, and the relationship of exposure to safety.11 Continued approval for this extended-dosing interval may be contingent upon verification and description of clinical benefit in the confirmatory trials.

Table 2. FDA-Approved CTLA-4 and PD-1 Inhibitors4-7
Cancer Types CTLA-4 Inhibitor PD-1 Inhibitors
Ipilimumab Nivolumab Pembrolizumab Cemiplimab
Adjuvant therapy
Melanoma
Unresponsive early stage disease
Non-muscle invasive bladder cancer    
Treatment of unresectable or metastatic disease
Classical Hodgkin lymphoma    
Cutaneous squamous cell carcinoma  
Melanoma
Merkel cell    
Urothelial    
Treatment of advanced disease as first-line therapy
Head & neck    
Melanoma    
MSI-high colon    
NSCLC    
Treatment of advanced disease as a second- or subsequent-line therapy
Cervical    
Classical Hodgkin lymphoma  
Esophageal (squamous cell)  
Gastric    
Head & neck (squamous cell)  
Hepatocellular  
Melanoma    
MSI-high cancer    
MSI-high colon  
NSCLC  
Primary mediastinal large B-cell lymphoma    
Renal cell    
SCLC    
TMB-H cancer    
Urothelial  
Abbreviations used: CTLA-4, cytotoxic T-lymphocyte antigen-4; FDA, U.S. Food and Drug Administration; MSI, microsatellite instability; NSCLC, non-small cell lung cancer: PD-1, programmed cell death-1; SCLC, small-cell lung cancer; TMB-H, tumor mutational burden high.
Table 3. FDA-Approved CTLA-4 and PD-1 Inhibitors in Combinations4-6
Cancer Types Combination Therapies
Ipilimumab + Nivolumab Pembrolizumab + Axitinib Pembrolizumab + Chemotherapy Pembrolizumab + Lenvatinib
Treatment of unresectable or metastatic disease as a first-line therapy
Head & neck    
Melanoma    
NSCLC a  
Pleural mesothelioma    
Renal  •  
TNBC    
Treatment of unresectable or metastatic disease as a second- or subsequent-line therapy
Endometrial cancer    
Hepatocellular    
Melanoma    
MSI-high colon    
Abbreviations used: CTLA-4; cytotoxic T-lymphocyte antigen-4; FDA, Food and Drug Administration; MSI, microsatellite instability; NSCLC, non-small cell lung cancer: PD-1, programmed cell death-1; TNBC, triple negative breast cancer.

a With or without platinum-doublet chemotherapy.
Table 4. FDA-Approved PD-L1 Inhibitors8-10
Cancers PD-L1 Inhibitors Combination Therapies
Atezolizumab Avelumab Durvalumab Atezolizumab + Chemotherapy Atezolizumab + cobimetinib and vemurafenib Avelumab + Axitinib Durvalumab + Chemotherapy
Maintenance Therapy
Stage III NSCLC            
Advanced urothelial            
Treatment of unresectable or metastatic disease
Breast cancer, triple negative            
Melanoma            
Merkel cell            
Urothelial            
Treatment of unresectable or metastatic disease as a first-line therapy
HCC a            
NSCLC          
Renal cell            
SCLC          
Treatment of unresectable or metastatic disease as second- or subsequent-line therapy
NSCLC            
Urothelial        
Abbreviations used: ALL, acute lymphoblastic leukemia; HCC, hepatocellular; NSCLC, non-small cell lung cancer; PD-L1, programmed cell death ligand-1; SCLC, small-cell lung cancer.

a Combined with bevacizumab
Table 5. FDA-Approved Checkpoint Inhibitors Initial- and Extended-Dosing Intervals4-10
Drugs Initial Checkpoint Inhibitors
Approved Dosing Regimens
Extended-interval Dosing Based on Modeling Studies
Atezolizumab 840 mg every 2 weeks, 1200 mg every 3 weeks 1680 mg every 4 weeksa
Avelumab 800 mg every 2 weeks
Cemiplimab 350 mg every 3 weeks None
Durvalumab 10 mg/kg every 2 weeks 1500 mg every 3 weeks, then 1500 mg every 4 weeksb
Ipilimumab 3 mg/kg every 3 weeks, 10 mg/kg every 3 weeks, 1 mg/kg every 3 weeks 1 mg/kg every 6 weeks (NSCLC, mesothelioma) 10 mg/kg every 3 weeks x 4 doses, followed by 10 mg/kg ever 12 weeks (Adjuvant melanoma) 1 mg/kg/ every 3 weeks x 4 doses) (metastatic melanoma advanced RCC, hepatocellular carcinoma, MSI-H colorectal cancer)
Nivolumab 240 mg every 2 weeks, 360 mg every 3 weeks 480 mg every 4 weeksc
Pembrolizumab 200 mg every 3 weeks 400 mg every 6 weeksd
Abbreviations used:
aWhen given as single agent.
bFor extensive-stage SCLC only 1500 mg every 3 weeks prior to etoposide + either carboplatin or cisplatin, then every 4 weeks as a single agent.
cFor all indications except mesothelioma.
dFor adult indications.

Continued March Toward Personalized Anticancer Therapy

In many but not all cancers, the tumors are associated with specific genetic mutations. Personalized anticancer therapy is based upon the premise that each patient’s tumor has a unique genetic profile, and therefore the concept that a single therapeutic option should be used for all patients and every tumor should be replaced with personalized anticancer treatments.12

Such personalized therapy allows for specific anticancer regimens to be used in patients with specific mutations. A good example of this is with advanced or metastatic non-small cell lung cancer (NSCLC). The treatment of NSCLC is dependent on the histologic subtype (i.e., the microscopic anatomy of the tumor) and molecular testing. The most common histologic NSCLC subtype is adenocarcinoma. Many of these tumors have genetic mutations, such as epidermal growth factor receptor (EGFR) mutations, anaplastic lymphoma kinase (ALK) gene rearrangements (also known as fusions), ROS1 rearrangements (also known as fusions), neurotrophic tyrosine receptor kinase (NTRK) gene fusions, BRAF mutations, c-mesenchymal-epithelial transition factor (c-MET) exon 14 skipping mutations, and rearranged during transfection (RET) fusions.13

Another concept that has begun to be incorporated into personalized anticancer therapy is the use of precision immunotherapy. This entails the optimal use of immunotherapy based upon the presence of predictive biomarkers for clinical response and new drug development that targets neo-antigens and mutational signatures of tumors.

Advanced NSCLC is also a good example of a condition in which precision immunotherapy is commonly used. Some tumors express PD-L1, indicating a more likely chance of responding to checkpoint inhibitors.12 Thus, the selection of therapy for a patient with advanced NSCLC of adenocarcinoma subtype is based upon the presence or absence of a mutation or PDL1 expression. The use of personalized therapy expands far beyond NSCLC, but is not as integral to all cancers as it is with advanced NSCLC.

Nonetheless, during 2020, many novel therapies were approved for personalized therapy indications as well as some that were already approved for previous indications but now have new indications (see Table 6).1

Table 6. Personalized Anticancer Agents and Indications Approved in 20201
Drug Indication
Targeted Therapy

Avapritinib (Ayvakit)

Unresectable or metastatic GIST harboring PDGFRA exon 18 mutation, including D842V mutations

Brigatinib (Alunbrig)

Anaplastic lymphoma kinase–positive metastatic NSCLC (now as first-line therapy rather than after progression or intolerance to crizotinib, as approved in 2017) and companion diagnostic Vysis Break Apart FISH probe kit

Capmatinib (Tabrecta)

NSCLC with c-MET exon 14 skipping as detected by an FDA-approved test

Encorafenib (Braftovi)

· Metastatic colorectal cancer with BRAF V600E mutation, detected by an FDA-approved test, after prior therapy (in combination with cetuximab; (2020)

· Prior approval (2017): Unresectable or metastatic melanoma with BRAF V600E or V600K mutation (in combination with binimetinib)

Neratinib (Nerlynx)

· Advanced or metastatic HER2-positive breast cancer who have received 2 or more prior anti-HER2 based regimens in the metastatic setting (in combination with capecitabine)

· Prior approval (2017): Extended adjuvant treatment of early-stage HER2 positive breast cancer following adjuvant trastuzumab therapy

Olaparib (Lynparza)

· Maintenance treatment of advanced epithelial ovarian, fallopian tube, or primary peritoneal cancer who are in complete or partial response to first-line platinum-based chemotherapy and whose cancer is associated with homologous recombination deficiency–positive status defined by either a deleterious or suspected deleterious BRCA mutation, and/or genomic instability in combination with bevacizumab, using FDA-approved companion diagnostic test

· Deleterious or suspected deleterious germline or somatic HRR gene–mutated mCRPC that has progressed following prior treatment with enzalutamide or abiraterone

· Prior approval (2014): Deleterious or suspected delirious germline BRCA-mutated advanced ovarian cancer who have been treated with 3 or more lines of chemotherapy, using FDA-approved companion diagnostic test

· Prior approval (2017): Maintenance of recurrent epithelial ovarian, fallopian tube, or primary peritoneal cancer who are in complete or partial response to platinum-based chemotherapy

· Prior approval (2018): Deleterious or suspected deleterious germline BRCA-mutated, HER-2 negative metastatic breast cancer previously treated with chemotherapy, using FDA-approved companion diagnostic test

· Prior approval (2018): Maintenance therapy for deleterious or suspected deleterious germline or somatic BRCA-mutated, epithelial ovarian, fallopian tube, or primary peritoneal cancer who are in complete or partial response to first-line platinum-based chemotherapy; using FDA-approved companion diagnostic test

· Prior approval (2019): Maintenance therapy for deleterious or suspected deleterious germline BRCA-mutated, metastatic pancreatic cancer, using FDA-approved companion diagnostic test

Pemigatinib (Pemazyre)

Adults with previously treated, unresectable, locally advanced or metastatic cholangiocarcinoma with a FGFR2 fusion or other rearrangement as detected by an FDA-approved test

Pralsetinib (Gavreto)

Metastatic RET fusion-positive NSCLC as detected by an FDA-approved test

Rucaparib (Rubraca)

· Deleterious BRCA mutation (germline and/or somatic) mCRPC in patients who have been treated with androgen receptor-directed therapy and a taxane-based chemotherapy

· Prior approval (2016): Deleterious BRCA-mutation (germline and/or somatic), epithelial ovarian, fallopian tube, or primary peritoneal cancer who have been treated with 2 or more chemotherapies; using an FDA-approved diagnostic test

· Prior approval (2018): Maintenance treatment of adult patients with epithelial ovarian, fallopian tube, or primary peritoneal cancer who are in complete or partial response to platinum-based chemotherapy

Selpercatinib (Retevmo)

· Metastatic RET fusion-positive NSCLC

· Advanced or metastatic RET-mutant medullary thyroid cancer (who require systemic therapy

· Advanced or metastatic RET fusion-positive thyroid cancer who require systemic therapy and who are radioactive iodine-refractory (if radioactive iodine is appropriate)

Tazemetostat (Tazverik)

Relapsed or refractory follicular lymphoma whose tumors are positive for an EZH2 mutation as detected by an FDA-approved test and who have received at least 2 prior systemic therapies; adult patients with relapsed or refractory follicular lymphoma who have no satisfactory alternative treatment options; adult and pediatric patients 16 and older with metastatic or locally advanced epithelioid sarcoma not eligible for complete resection

Tucatinib (Tukysa)

In combination with trastuzumab and capecitabine for treatment of adult patients with advanced unresectable or metastatic HER2-positive breast cancer, including patients with brain metastases, who have received 1 or more prior anti-HER2-based regimens in the metastatic setting

Monoclonal Antibodies

Gemtuzumab ozogamicin (Mylotarg)

· Newly diagnosed CD33-postive acute myeloid leukemia pediatric patients

· Prior approval (2017): Newly diagnosed CD33-positive acute myeloid leukemia

· Prior approval (2017): Relapsed/refractory CD33-positive acute myeloid leukemia

Margetuximab-cmkb (Margenza)

· Adults with metastatic HER2-positive breast cancer who have received 2 or more prior anti-HER2 regimens, at least 1 of which was for metastatic disease in combination with chemotherapy

Naxitamab-gqgk (Danyelza)

· Adult and pediatric patients 1 year of age or older with relapsed or refractory high-risk neuroblastoma in the bone or bone marrow demonstrating a partial response, minor response, or stable disease prior to therapy in combination with GM-CSF

Ramucirumab (Cyramza)

· First-line treatment of metastatic NSCLC with EGFR exon 19 deletions or exon 21 (L858R) mutations in combination with erlotinib

· Prior approval (2014): Metastatic NSCLC with disease progression on/after platinum-based chemotherapy or those with EGFR or ALK mutations after FDA-approved therapies for these mutations (previously approved in combination with docetaxel)

· Prior approval (2015): Advanced or metastatic gastric or gastro-esophageal junction adenocarcinoma with disease progression on/after fluoropyrimidine (alone or in combination with paclitaxel)

· Prior approval (2015): Metastatic colorectal cancer with disease progression on/after bevacizumab, oxaliplatin, and fluoropyrimidine (with FOLFIRI)

· Prior approval (2019): Hepatocellular carcinoma with alpha fetoprotein ≥400 ng/mL and treated with sorafenib

Sacituzumab govitecan-hziy (Trodelvy)

Metastatic triple-negative breast cancer who received at least 2 prior therapies for metastatic disease

Immunotherapya

Atezolizumab (Tecentriq)

· First-line treatment of adult patients with NSCLC whose tumors have high PD-L1 expression (PD-L1 stained ≥50% of tumor cells or PD-L1 stained tumor-infiltrating immune cells covering ≥10% of the tumor area), with no EGFR or ALK genomic tumor aberrations

· BRAF V600 mutation-positive unresectable or metastatic melanoma (iin combination with cobimetinib and vemurafenib)

· In combination with paclitaxel protein-bound for the treatment of adult patients with unresectable locally advanced or metastatic TNBC whose tumors express PD-L1

· In combination with bevacizumab for the treatment of patients with unresectable or metastatic hepatocellular carcinoma who have not received prior systemic therapy

Nivolumab (Opdivo), + Ipilimumab (Yervoy)

· First-line treatment for patients with metastatic NSCLC whose tumors express PD-L1 (≥1%), as determined by an FDA-approved test, with no EGFR or ALK genomic tumor aberrations

· Nivolumab + ipilimumab + 2 cycles of platinum-doublet chemotherapy as first-line treatment for patients with metastatic or recurrent NSCLC, with no EGFR or ALK genomic tumor aberrations

· First-line treatment in adult patients with unresectable malignant pleural mesothelioma

Pembrolizumab (Keytruda)

· Treatment of patients with locally recurrent unresectable or metastatic triple-negative breast cancer whose tumors express PD-L1 (combined positive score ≥10) as determined by an FDA-approved test

· Adult patients with relapsed or refractory classical Hodgkin lymphoma

· Pediatric patients with refractory classical Hodgkin lymphoma or that has relapsed after 2 or more lines of therapy

· Treatment of adult and pediatric patients with unresectable or metastatic TMB-H (≥10 mutations/megabase) solid tumors, as determined by an FDA-approved test, that have progressed following prior treatment and who have no satisfactory alternative treatment options

· First-line treatment of patients with unresectable or metastatic microsatellite instability-high or mismatch repair deficient colorectal cancer

Abbreviations used: ALK, anaplastic lymphoma kinase; CD, cluster of differentiation; FDA, U.S. Food and Drug Administration; FGFR, fibroblast growth factor receptor; GIST, gastrointestinal stromal tumor; GM-CSF, granulocyte macrophage colony stimulating factor; HRR, homologous recombination repair; MEK, mitogen-activated protein kinase; mCRPC, metastatic castration-resistant prostate cancer; NSCLC, non-small cell lung cancer; PDGFRA, platelet-derived growth factor receptor A; TMB H, tumor mutational burden–high.

aFor immunotherapy agents, many other pre-existing indications exist (see Tables 2, 3 and 4).

Pembrolizumab was the only anticancer agent to have a tumor-agnostic indication approved in 2020.1 Tumor-agnostic therapies are drugs used to treat cancer based on the cancer’s genetic and molecular features without regard to the cancer type. That is, tumor-agnostic drugs can be used for any cancer that has that particular genetic and molecular feature.14 To date, only 3 therapies have this designation: pembrolizumab, larotrectinib, and entrectinib. It is expected that many more of these tumor-agnostic indications will be approved in the future.

In 2017, the PD-1 inhibitor pembrolizumab became the first drug to receive this approval for the treatment for adult and pediatric patients with unresectable or metastatic, microsatellite instability-high (MSI-H), or mismatch repair deficient (dMMR) solid tumors that have progressed following prior treatment and who have no suitable alternative treatment options. In 2020, it received its second tumor-agnostic indication: adult and pediatric patients with unresectable or metastatic tumor mutational burden-high (TMB H) [≥10 mutations/megabase (mut/Mb)] solid tumors.1,6

Larotrectinib (Vitrakvi) is an oral tyrosine kinase inhibitor that specifically inhibits tropomyosin receptor (TRK) kinase.14,15 It was approved in November 2018 for treatment of adult and pediatric patients with solid tumors that have a neurotrophic TRK gene fusion in the metastatic setting or for whom surgical resection is not an option.

Entrectinib (Rozlytrek) is another TRK inhibitor that received approval in August 2019 for the same population as larotrectinib.16 In addition to these tumor-agnostic indications, entrectinib also has tumor-specific indications.16

Disease Updates: Bladder and Small-Cell Lung Cancer

Bladder Cancer

Big advancements in both bladder and small-cell lung cancer occurred in 2020. Historically, systemic therapy has not been used for treatment of early bladder or upper urinary tract cancers.1,17 In fact, until 2020, all of the anticancer treatments approved had been for advanced or metastatic disease.1

In January 2020, pembrolizumab (Keytruda) became the first systemic therapy approved for early-stage bladder cancer.1 Bladder cancer is the sixth most common cancer diagnosed in the United States.17,18 It typically is a disease of older patients, with the median age of diagnosis of 73 years. The most common form of bladder cancer is nonmuscle invasive bladder cancer (NMIBC), accounting for 70% of bladder cancers.19 It has 5-year survival rate of more than 88%, but these tumors typically recur after initial treatment, and 10% to 20% can progress to the more advanced form, muscle-invasive bladder cancer (MIBC). The management of NMIBC, therefore, is aimed at reducing recurrences and preventing progression to MIBC.17 In fact, these high rates of progression and recurrent with current therapy require life-long surveillance.

The typical treatment of NMIBC is a transurethral resection of the bladder tumor (TURBT) with or without intravesical therapy with Bacillus Calmette-Guerin (BCG) or chemotherapy (e.g., mitomycin, gemcitabine). In patients who are unresponsive to BCG and have high-risk disease with carcinoma in situ (CIS) with or without papillary tumors who are ineligible for or have elected not to undergo cystectomy, pembrolizumab is a new treatment option. Efficacy and safety data come from the KEYNOTE-057 study, which was a multicenter, single-arm study evaluating 148 patients with high-risk NMIBC. They received pembrolizumab 200 mg intravenously every 3 weeks for 24 months; 41% of the 96 patients with BCG-unresponsive CIS with or without papillary tumors had a complete response lasting for a median of 16.2 months. Overall, this therapy was very well tolerated, with fatigue occurring in about 30% of patients. Immune-mediated adverse events requiring discontinuation occurred in only 11% of patients.6

In April 2020, mitomycin for pyelocalyceal solution (Jelmyto) was approved for the treatment of adult patients with low-grade upper tract urothelial cancer.1 Mitomycin, a cytotoxic drug that inhibits the synthesis of DNA, RNA, and proteins, was first approved in 1974 for the treatment of advanced cancer of the stomach or pancreas but has been used off-label for several other cancers, such as anal, bladder, cervical, esophageal, hepatocellular, or vulvar cancers.20,21 With newer, more effective drugs for each of these cancers, mitomycin is not used often. This new formulation of mitomycin is a ureteral gel (not an intravenously administered drug) and is the first therapy approved by FDA for upper tract urothelial cancer. Based on recent findings, it is now considered the standard of care following complete or near complete resection of upper tract urothelial tumors (e.g., tumors that originate in the renal pelvis or ureter).17,22

These upper tract urothelial tumors are quite rare and up until 2020, the primary treatment was surgery alone.17 For patients with low-grade tumors, the primary treatment is an endoscopic resection or nephroureterectomy (removal of kidney, entire ureter, and a small piece of bladder with the ureter and bladder connect). For high-grade tumors, neoadjuvant chemotherapy followed by nephroureterectomy is preferred. Because low-grade tumors often recur, adding mitomycin gel following complete resection or ablation of the tumor improves disease outcomes.17

The OLYMPUS study, a single-arm, multicenter, phase 3 study showed that 6 weekly instillations (via pyelocaliceal application and can be administered via ureteral catheter or a nephrostomy tube) of mitomycin ureteral gel into the renal pelvis and calyces produced a durable response in 84% of patients at 12 months.23 Overall, this drug is well tolerated; however, ureteric stenosis, urinary tract infections, hematuria, flank pain, and nausea did occur in more than 20% of patients.

This gel formulation of mitomycin has some unique administration considerations. Several steps are involved in its preparation, including using a chilling block and determining the volume to be administered based on the patient’s kidney volume.22 Having the patient take 1.3 grams of sodium bicarbonate orally the evening before, morning of, and 30 minutes before improves efficacy.

In May 2020, the interim results of the JAVELIN Bladder 100 study were presented at the annual American Society of Clinical Oncology meeting; these led to significant changes in the way clinicians manage metastatic MIBC.17,24 Metastatic MIBC is not curable; therefore, the goal of therapy is to prolong life while maintaining quality of life.17

The JAVELIN Bladder 100 study included 700 patients with complete or partial response or stable disease after first-line platinum-based chemotherapy for metastatic MIBC; they were randomized to the PD-L1 inhibitor avelumab 10 mg/kg intravenously every 2 weeks plus best supportive care or best supportive care alone.24,25 Until now, maintenance therapy was not part of the management of metastatic MIBC. The results of this study, however, showed that maintenance therapy with avelumab improved overall survival regardless of PD-L1 status (21.4 mo vs 14.3 mo; hazard ratio [HR], 0.69; 95% confidence interval [CI] 0.56–0.86; P <0.001). Avelumab was well tolerated with only 11% of patients experiencing treatment-related events. Avelumab is now part of the standard of care as maintenance therapy following platinum-based chemotherapy. It is continued until disease progression or treatment-related adverse effects require discontinuation.17

Small-Cell Lung Cancer

In the United States, lung cancer remains the leading cause of cancer-related deaths in both men and women.18 Lung cancers can be categorized as non-small cell lung cancer, small cell lung cancer (SCLC), and primary neuroendocrine tumors. Approximately 10% to 15% of all lung cancers are in the SCLC category; these are aggressive tumors often characterized by rapid growth, leading to early metastatic spread but also initially very sensitive to chemotherapy and radiation therapy.26 Almost all SCLC cases are attributable to smoking.

Most patients are diagnosed with advanced SCLC, often referred to as extensive-stage SCLC. The goal of treatment in these patients is to improve survival while maintaining quality of life. Until 2018, the standard treatment for extensive-stage SCLC had remained the same for 20 years: 4 to 6 cycles of a platinum (either carboplatin or cisplatin) + etoposide chemotherapy.26 Treatment for relapsed extensive-stage SCLC depended on timing of the relapse, and outcomes with single-agent chemotherapy were dismal. Nivolumab and pembrolizumab were found to be effective in relapsed disease in 2018 and 2019, respectively.5,6 Since those approvals, checkpoint inhibitor therapy is being used in combination with 4 cycles of platinum/etoposide chemotherapy and is then continued as maintenance therapy.26

In 2020, further advancements in the treatment of SCLC occurred, continuing to change the way this disease is treated. In March 2020, durvalumab was approved as first-line therapy in combination with platinum/etoposide for 4 cycles followed by durvalumab maintenance therapy. 1 This was based on the results of the CASPIAN trial, a randomized, phase 3 trial comparing this regimen to platinum/etoposide alone.27 The overall survival time with durvalumab plus chemotherapy was 13.0 months compared to 10.3 months with platinum plus etoposide alone, for a HR of 0.73 (95% CI, 0.59–0.91; P = 0.0047). These data provide a choice for providers in terms of the first-line combinations and maintenance therapies.

In March 2019, the FDA approved atezolizumab as first-line therapy of SCLC in combination with 4 cycles of carboplatin and etoposide and continued as maintenance based on the results of the IMpower133 study.28 Therefore, providers can choose either this regimen or the durvalumab + cisplatin or carboplatin plus etoposide followed by durvalumab maintenance therapy. No head-to-head comparisons have been done so far.

A new cytotoxic agent lurbinectedin (Zepzelca) was approved in June 2020 for the treatment of extensive-stage SCLC that has progressed on or after platinum-based chemotherapy.1 Lurbinectedin is a cytotoxic agent, an alkylating drug that binds to guanines in DNA.29-31 This results in the formation of adducts, bending the DNA helix, thereby interrupting the cell cycle and producing apoptosis.

Results of a single-arm, open-label, phase 2 basket trial showed that lurbinectedin 3.2 mg/m2 intravenously every 3 weeks was effective as a second-line therapy in extensive-stage SCLC patients.29 In this study, a 35.2% of patients had an overall response, with most partial responses and stable disease, with a median duration of response of 5.3 months (95% CI, 4.1-6.4). Although this response rate may not seem very optimistic, it is indeed noteworthy in the second-line treatment setting for extensive-stage SCLC, particularly in patients who are resistant to platinum-based therapy. To date, the only evidence-based second-line therapy is topotecan, with overall response rates of 16%.26

Lurbinectedin is overall safe with an acceptable toxicity profile. Reversible myelosuppression is the most common adverse event. No treatment-related deaths were observed. As a result of this trial, the NCCN guidelines for treatment of extensive-stage SCLC recommend either topotecan, lurbinectedin, or enrollment in a clinical trial for patients who relapse within 6 months of first-line therapy.26

Monoclonal Antibodies – Moving Toward Subcutaneous Administration

Monoclonal antibodies are widely used in the treatment of both solid tumors and hematologic malignancies and are mainly given intravenously.32 This route of administration often requires several hours because of the risk of infusion-related reactions. Therefore, the appeal of subcutaneous administration is not only shorter visits for the patient, but also a less invasive administration method that creates the possibility of self-administration. Challenges include reducing pain when with larger fluids volumes sometimes necessary to deliver the appropriate dose, ensuring good absorption and bioavailability, and appropriate technique to ensure exact doses are administered.

Subcutaneous administration of biologics has been well established in noncancer disease states, such as multiple sclerosis and rheumatoid arthritis for many years.32 In 2017, the first oncology monoclonal antibody for subcutaneous administration was approved, rituximab and hyaluronidase human (Rituxan Hycela).1 A similar trastuzumab product was approved in 2019, trastuzumab and hyaluronidase-oysk injection (Herceptin Hylecta). In 2020, 2 more subcutaneous monoclonal antibodies were approved: daratumumab + hyaluronidase-fihj (Darzalex Faspro) for adult patients with newly diagnosed or relapsed/refractory multiple myeloma and a fixed-dose combination of pertuzumab, trastuzumab, and hyaluronidase–zzxf (Phesgo). The hyaluronidase component of these products improves absorption within the skin.33 It is important to note that these drugs are not completely interchangeable with similar intravenous products, and they are not intended for home self-administration because of the risk of reactions and in some cases complicated doses/administration (see Table 7).33-36

Table 7. Monoclonal Antibodies for Subcutaneous Administration33-36
Drug Indication Notes

Daratumumab + hyaluronidase-fihj

(Darzalex Faspro)

Multiple myeloma

· In combination with bortezomib, melphalan, and prednisone in newly diagnosed patients who are ineligible for autologous HSCT

· In combination with lenalidomide and dexamethasone in newly diagnosed patients who are ineligible for autologous HSCT and in patients with relapsed or refractory multiple myeloma who have received at least 1 prior therapy

· In combination with bortezomib and dexamethasone in patients who have received at least 1 prior therapy

· As monotherapy, in patients who have received at least 3 prior lines of therapy including a proteasome inhibitor and an immunomodulatory agent or who are double refractory to a proteasome inhibitor and an immunomodulatory agent

· Fixed dose (i.e., different dose), withdrawn from a single-dose vial; not a prefilled syringe

· Volume = 17 mL

· Administered SC (alternate between left and right navel) over 3–5 min

· Requires premedication (1–3 hr before administration) with H1 receptor antagonist, acetaminophen, and corticosteroid; recommended observation after 1st and 2nd dose; postmedication with corticosteroids may be needed to decrease risk of delayed administration reactions

· Must be administered by a health care professional

Pertuzumab, trastuzumab, and hyaluronidase–zzxf (Phesgo)

Use in combination with chemotherapy as:

· Neoadjuvant treatment of patients with HER2-positive, locally advanced, inflammatory, or early-stage breast cancer (either greater than 2 cm in diameter or node positive) as part of a complete treatment regimen for early breast cancer

· Adjuvant treatment of patients with HER2-positive early breast cancer at high risk of recurrence

Use in combination with docetaxel for treatment of patients with HER2-positive metastatic breast cancer who have not received prior anti-HER2 therapy or chemotherapy for metastatic disease

· Fixed dose (i.e., different dose) that has both an initial fixed dose and a maintenance fixed dose

· Withdrawn from single-dose vial; not a prefilled syringe

· Must be administered by a health care professional, who must observe the patient for 30 minutes after initial dose and 15 minutes after maintenance doses

· If in combination with anthracycline, administered after anthracycline

· If in combination with docetaxel or paclitaxel, administer before docetaxel or paclitaxel

· Volume = 15 mL (initial dose); 10 mL (maintenance dose)

· Administered initial dose SC (in thigh only) over 8 min; maintenance dose over 5 min

· Do not substitute for or with ado-trastuzumab emtansine

· Patients receiving IV pertuzumab and trastuzumab can transition; if it <6 wk, administered maintenance dose; if >6 wk, administered initial dose then maintenance dose

Rituximab + hyaluronidase (Rituxan Hycela)

· Follicular lymphoma (as single agent in relapsed/refractory; previously untreated in combination with first-line chemotherapy and in those with CR or PR to rituximab with combination chemotherapy as a single agent; nonprogressing type as single agent after CVP)

· Diffuse large-B cell lymphoma (previously treated in combination with CHOP or other anthracycline-based chemotherapy)

· Chronic lymphocytic leukemia (previously untreated or treated) in combination with fludarabine and cyclophosphamide

· Must receive 1 full dose of rituximab with side effects before initiating

· Fixed dose (i.e., different dose), withdrawn from single-dose vial; not a prefilled syringe

· Dose (and volume) varies based on indication

· Requires premedications (acetaminophen, antihistamine; consider glucocorticoids) and observation of patient for 15 minutes after injection

· Administered SC in abdomen over 5–7 min

· Must be administered by a health care professional

· Not indicated for nonmalignant conditions

Trastuzumab + hyaluronidase-oysk (Herceptiin Hylecta)

· HER2+ breast cancer

· Fixed dose (i.e., different dose), withdrawn from single-dose vial; not a prefilled syringe

· Volume = 5 mL

· Administered SC (alternate between left and right thigh) over 2–5 min

· Do not substitute for or with ado-trastuzumab emtansine

· Approved based on noninferiority and safety study

· Should be administered by a health care professional

Abbreviations used: CHOP, cyclophosphamide, doxorubicin, vincristine, prednisone; CR, complete response; CVP, cyclophosphamide, vincristine, prednisone; HSCT, hematopoietic stem cell transplantation; IV, intravenous; PR, partial response; SC, subcutaneous.

COVID-19 and Cancer

During the pandemic of coronavirus disease 2019 (COVID-19), older adults and those with severe underlying medical conditions (e.g., heart disease, chronic obstructive pulmonary disease, type 2 diabetes) or cancer have had a higher likelihood of morbidity and mortality after developing symptoms from the causative virus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).37 In fact, several retrospective studies have been undertaken to learn more about those with cancer who had COVID-19.38-40 Some of the factors that been linked include a higher incidence in males than females and similar to the normal population, it occurs more commonly in older adults and those with comorbidities.38

No one type of cancer seems to cause an increased risk of developing COVID-19. However, those with recent exposure to the health care system were more likely to have an increased risk of COVID-19 infection and subsequently severe outcomes (e.g., intubation, death). In patients with COVID-19 treated in the United States, Canada, and Spain, COVID-19 resulted in hospitalization in 40% of patients with cancer, intubation in 12%, and death in 13%. The 30-day mortality rate was higher in older adults (odds ratio [OR], 1.84), those that were male (OR, 1.63), had 2 or more comorbidities (OR, 4.5) and a poorer performance status (Eastern Cooperative Oncology Group performance status: 2 or higher; OR, 3.89).39

The third and largest study was conducted by Memorial Sloan Kettering Cancer Center, evaluating 2,035 patients with cancer tested between March 10 and April 7, 2020.40 Of these patients, 423 (21%) were diagnosed with symptomatic COVID-19, 40% of whom were hospitalized and 20% had severe respiratory illness. Nine percent of patients were ventilated and 12% died within 30 days of diagnosis. Risks associated with hospitalization included nonwhite race, hematologic malignancy, chronic lymphopenia and/or corticosteroid use, and immune checkpoint inhibitor therapy. Risks associated with severe respiratory illness included age >65 years and immune checkpoint inhibitor therapy. Interestingly, chemotherapy within the 30 days prior to COVID-19, surgery, and metastatic disease were not associated with a higher risk of poor outcomes. The rationale for risk associated with immune checkpoint inhibitor therapy is unclear. Some theories include treatment-related lung injury, corticosteroid use for immune-related adverse events; or an immune checkpoint inhibitor–triggered immune dysregulation by T-cell hyperactivation, leading to acute respiratory distress syndrome.

As a result of this information about patients with cancer and the need to reduce the rates of COVID-19, best practices have quickly emerged in caring for patients with cancer. The American Society of Clinical Oncology (ASCO) has provided some guidance for management of cancer patients during the pandemic (see Table 8).41 As the COVID-19 pandemic continues and clinicians learn more about how to best manage patients, new information is likely to become available.

Some other great COVID-19 resources for patients with cancer can be found on the Hematology/Oncology Pharmacy Association’s COVID-19 website: https://www.asco.org/asco-coronavirus-resources/care-individuals-cancer-during-covid-19/cancer-treatment-supportive-care, which has links to multiple organizations that are keeping up to date with the latest best practices.

Table 8. ASCO’s Guidance on COVID-19 in Patients With Cancer 41
Cancer Treatment and Supportive Care Situation Recommendation

Stop, halt or delay current therapy?

Decisions about interrupting anticancer treatment in patients with active COVID-19 should be based on a clinical benefit: risk assessment that considers risk of interrupting cancer treatment versus poorly defined risk of adverse COVID-19 outcomes in patients receiving active cancer treatment

COVID-19 testing

Priority 1:

· Hospitalized patients with symptoms

· Symptomatic residents of long-term care facilities or other congregate living settings, including prisons and shelters

Priority 2:

· Persons with COVID-19 symptoms

Priority 3:

· Asymptomatic patients prior to immunosuppressive therapy (e.g., cytotoxic chemotherapy, stem cell transplantation, biologic therapy, cellular immunotherapy, or high-dose corticosteroids) within 48–72 hr of 1st dose

· Asymptomatic individuals prioritized by health departments or clinicians

Delay therapy in COVID-19+ patients?

Unclear how long a delay after SARS-CoV-2 infection has resolved may be necessary before initiating/restarting anticancer therapy, but if the decision was made to interrupt treatment, it should not be resumed until COVID-19 symptoms have resolved and clinicians are reasonably certain that virus is no longer present (e.g., a negative SARS-CoV-2 test), unless cancer is rapidly progressing and the risk–benefit assessment favors proceeding with cancer treatment

Therapy for infection

Prophylaxis: No evidence to support use of prophylactic antiviral therapy for COVID-19 in immunosuppressed patients

Treatment: U.S. National Institutes of Health has recommendations for infection treatment, including the use of remdesivir and dexamethasone; recommendations are changing rapidly and should be reviewed prior to considering therapy

Neutropenic fever

Prophylaxis: Reasonable for patients at risk for neutropenic fever to receive colony-stimulating growth factor for treatment regimens at a lower level of expected risk (e.g., >10% risk) to minimize risk of neutropenic fever and the potential need for emergency care, with instructions for neutrophil count monitoring and regular contact with their health care team

· Acute care for potential neutropenic fever: Consider evaluation by telemedicine or by phone to determine whether a patient should be evaluated in the clinic or sent to the emergency department

· Acute care for known neutropenic fever: Follow standard guidelines for care of neutropenic patients with fever, including isolation; rapid COVID-19 testing should be used, if available, to determine personal protective equipment necessary for caregivers and appropriate location in facility for continued care. In absence of rapid testing, manage patient for neutropenic fever per standard guidelines, presuming COVID-19 infection.

Cancer-related anemia

· Prophylaxis: Consider erythropoietin-stimulating agents when serious and/or symptomatic cancer/treatment-related anemia is anticipated and agents are deemed safe; prophylactic transfusion in asymptomatic patients based on laboratory values should be avoided.

· Acute care: Transfusion should be given when serious and/or symptomatic cancer/treatment-related anemia occurs in accordance with usual practice. The American Society of Hematology previously recommended not transfusing more than minimum number of red blood cell units necessary to relieve anemia symptoms or to return patient to safe hemoglobin range (7 to 8 g/dL in stable, noncardiac inpatients). As blood donations may be affected by community public health measures, local blood supply must be considered as part of decision-making. Consider simultaneously initiating erythropoietin-stimulating agents when deemed safe.

Central venous catheters

Flushing can occur at frequencies as long as every 12 weeks with no notable increase in adverse events or harms; consider having patients flush their own devices but also that process of training may itself be a source of exposure and access to sterile supplies at home may be limited.

Abbreviations used: COVID-19, coronavirus disease 2019; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.

Conclusion

Many advancements were made in oncology during 2020. It can be difficult to stay abreast of the evolving treatment options for each type of cancer. However, because pharmacists are instrumental in the management of patients undergoing treatment for cancer, it is important to stay up to date.

More than 20 new tyrosine kinase inhibitors and cytotoxic agents were approved in 2020 as well as 30 additional indications for existing treatments. Extended-dosing intervals for immune checkpoint inhibitors have allowed patients to receive immunotherapy less frequently in some cases, potentially minimizing their risk of exposure to COVID-19. Differences exist in monoclonal antibodies that are available both in intravenous and subcutaneous formulations, and it is important for the pharmacist to be aware of these differences. Advancements in both bladder cancer and SCLC will allow more patients to receive systemic treatment and have better outcomes. Finally, as researchers and clinicians learn more about COVID-19 and cancer, it will be important for pharmacists to remain familiar with the latest guidance on how to manage patients with cancer while SARS-CoV-2 is circulating.

REFERENCES

  1. U.S. Food and Drug Administration. Hematology/Oncology (Cancer) approvals and safety notifications. Accessed November 9, 2020. https://www.fda.gov/drugs/resources-information-approved-drugs/hematologyoncology-cancer-approvals-safety-notifications
  2. Robert C. A decade of immune-checkpoint inhibitors in cancer therapy. Nat Commun. 2020;11:3801. https://doi.org/10.1038/s41467-020-17670-y.
  3. Renrick AR, Dunbar ZT, Shanker A. Update on the current revolution in cancer immunotherapy. 2019;11:15;20.
  4. Yervoy [ipilimumab] [package insert]. Princeton, NJ: Bristol-Myers Squibb Company; 2020.
  5. Opdivo [nivolumab] [package insert]. Princeton, NJ: Bristol-Myers Squibb Company; 2020.
  6. Keytruda [pembrolizumab] [package insert]. Whitehouse Station, NJ: Merck & Co, Inc.; 2020.
  7. Libtayo [cemiplimab] [package insert]. Tarrytown, NY: Regeneron Pharmaceuticals, Inc.; 2018.
  8. Tecentriq [atezolizumab] [package insert]. South San Francisco, CA: Genentech, Inc.; 2020.
  9. Bavencio [avelumab] [package insert]. Rockland, MA: EMD Serono, Inc.; 2020.
  10. Imfinzi [durvalumab] [package insert]. Wilmington, DE: AstraZeneca Pharmaceuticals, LP.; 2020.
  11. Sehgal K, Costa DB, Rangachari D. Extended-interval dosing strategy of immune checkpoint inhibitors in lung cancer: will it outlast the COVID-19 pandemic. Front Oncol. 2020;23 June 2020. https://doi.org/10.3389/fonc.2020.01193
  12. Mandal R, Chan TA. Personalized oncology meets immunology: the path toward precision immunotherapy. Cancer Discov. 2016;6(7):703-713.
  13. National Comprehensive Cancer Network (NCCN) Clinical Practice Guidelines in Oncology. Non-Small Cell Lung Cancer. Version 2.2021. Accessed January 16, 2020. https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf
  14. Looney A-M, Nawaz K, Webster RM. Tumor agnostic therapies. Nature. 2020;19:383-384.
  15. Vitrakvi [larotrectinib] [package insert]. Stamford, CT: Loxo Oncology, Inc.; 2018.
  16. Rozlytrek [entrectinib] [package insert]. South San Francisco, CA: Genentech, Inc.; 2019.
  17. National Comprehensive Cancer Network Guidelines Bladder Cancer Version 6. 2020. Accessed November 11, 2020. https://www.nccn.org/professionals/physician_gls/pdf/bladder.pdf
  18. Siegal RL, Miller KD, Jemal A. Cancers statistics, 2020. CA Cancer J Clin. 2020;70:7-30.
  19. Ryan K, Hale GR, Pena-La Grave G, et al. New therapies in nonmuscle invasive bladder cancer treatment. Indian J Urol. 2018;34:11-19.
  20. Mitozytrex [package insert]. Dublin, CA: SuperGen, Inc.; 2002.
  21. Lexicomp Online Database. Hudson (OH): Lexicomp Inc.: 2020.
  22. Jelmyto [package insert]. Princeton, NJ: UroGen Pharma, Inc.; April 2020.
  23. Kleinmann N, et al. Primary chemoablation of low-grade upper tract urothelial carcinoma using UGN-101, a mitomycin-containing reverse thermal gel (OLYMPUS): an open-label, single-arm, phase 3 trial. Lancet Oncol. 2020;21:776-785.
  24. Powles T, Park SH, Voog E, et al. Maintenance avelumab + best supportive care (BSC) versus BSC alone after platinum-based first-line (1L) chemotherapy in advanced urothelial carcinoma (UC): JAVELIN Bladder 100 phase III interim analysis. J Clin Oncol. 2020;38(18 suppl):LBA1.
  25. Powles T, Park SH, Voog E, et al. Avelumab maintenance therapy for advanced or metastatic urothelial carcinoma. N Engl J Med. 2020;383:1218-1230.
  26. National Comprehensive Cancer Network Guidelines Small Cell Lung Cancer Version 2.2021. Accessed January 16, 2020. https://www.nccn.org/professionals/physician_gls/pdf/sclc.pdf
  27. Paz-Ares L, Dvorkin M, Chen Y, et al. Durvalumab plus platinum-etoposide versus platinum-etoposide in first-line treatment of extensive-stage small-cell lung cancer (CASPIAN): a randomised, controlled, open-label, phase 3 trial. Lancet. 2019;394(10212):1929‐1939.
  28. S. Food and Drug Administration. FDA approves atezolizumab for extensive-stage small cell lung cancer. Accessed June 10, 2020. https://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-atezolizumab-extensive-stage-small-cell-lung-cancer
  29. Zepzelca [package insert]. Palo Alto, CA: Jazz Pharmaceuticals, Inc.; 2020.
  30. Trigo J, Subbiah V, Besse B, et al. Lurbinectedin as second-line treatment for patients with small-cell lung cancer: a single-arm, open-label, phase 2 basket trial. Lancet. 2020;21:645-654.
  31. Perez JMT, Leary A, Besse B, et al. Efficacy and safety of lurbinectedin (PM1183) in small cell lung cancer (SCLC): results from a phase 2 study. J Clin Oncol. 2018;36(suppl, abst 161909).
  32. Jackish C, Muller V, Maintz C, et al. Subcutaneous administration of monoclonal antibodies in oncology. Geburtshilfe Fraeunheikd. 2014;74:343-349.
  33. Herceptin Hylecta [package insert]. South San Francisco, CA: Genentech, Inc; 2020.
  34. Darzalex faspro [package insert]. Horsham, PA: Janssen Biotech, Inc.; 2020.
  35. Phesgo [package insert]. South San Francisco, CA: Genentech, Inc.; 2020.
  36. Rituxan Hycela [package insert]. South San Francisco, CA: Genentech, Inc.; 2020.
  37. Centers for Disease Control and Prevention. Symptoms of Coronavirus. 2020. Accessed November 11, 2020. https://www.cdc.gov/coronavirus/2019-ncov/symptoms-testing/symptoms.html
  38. Fung M, Babik JM. COVID-19 in immunocompromised hosts: what we know so far. Clin Infect Dis. 2021;72:340-350.
  39. Kuderer NM, Choueiri TK, Shah DP, et al. Clinical impact of COVID-19 on patients with cancer (CCC19): a cohort study. 2020;395(10241):1907-1918.
  40. Robilotti EV, Babady NE, Mead PA, et al. Determinants of COVID-19 disease severity in patients with cancer. Nat Med. 2020;26(8):1218-1223.
  41. American Society of Clinical Oncology. Cancer treatment & supportive care. Accessed November 11, 2020. https://www.asco.org/asco-coronavirus-resources/care-individuals-cancer-during-covid-19/cancer-treatment-supportive-care.

Back to Top