Cancer drugs are a central part of cancer therapy and knowledge of their use, mode of action, and side effect profile is very important. This chapter is intended to provide an overview and to serve as a reference. With this intent, Table 54.1 lists the acronyms and components of a number of combination therapies and Tables 54.2 to 54.12 summarize the information for a number of cancer therapeutics organized by drug class.

TABLE 54.1
Acronyms of Chemotherapy Regimens
NAME COMPONENTS EXAMPLE OF USES, AND OTHER NOTES
7+3, also known as DA or DAC in case of daunorubicin, or IA or IAC in case of idarubicin use 7 days of ara-C (cytarabine) plus 3 days of an anthracycline antibiotic, either daunorubicin [DA or DAC variant] or idarubicin [IA or IAC variant] Acute myelogenous leukemia, excluding acute promyelocytic leukemia
ABVD doxorubicin (Adriamycin), bleomycin, vinblastine, dacarbazine Hodgkin lymphoma
AC doxorubicin (Adriamycin), cyclophosphamide Breast cancer
BACOD bleomycin, doxorubicin (Adriamycin), cyclophosphamide, vincristine (Oncovin), dexamethasone Non-Hodgkin lymphoma
BEACOPP bleomycin, etoposide, doxorubicin (Adriamycin), cyclophosphamide, vincristine (Oncovin), procarbazine, prednisone bleomycin, etoposide, doxorubicin (Adriamycn), cyclophosphamide, vincristine (Oncovin), procarbazine, prednisone) Hodgkin lymphoma
BEP bleomycin, etoposide, platinum agent Testicular cancer, germ cell tumors
CA cyclophosphamide, doxorubicin (Adriamycin) (same as AC) Breast cancer
CAF cyclophosphamide, doxorubicin (Adriamycin), fluorouracil (5-FU) Breast cancer
CAPOX or XELOX capecitabine and oxaliplatin Colorectal cancer
CAV cyclophosphamide, doxorubicin (Adriamycin), vincristine Lung cancer
CBV cyclophosphamide, BCNU (carmustine), VP-16 (etoposide) Lymphoma
CHOEP cyclophosphamide, hydroxydaunorubicin (doxorubicin), etoposide, vincristine (Oncovin), prednisone Non-Hodgkin lymphoma
CEPP cyclophosphamide, etoposide, procarbazine, prednisone Non-Hodgkin lymphoma
ChlVPP/EVA chlorambucil, vincristine (Oncovin), procarbazine, prednisone, etoposide, vinblastine, doxorubicin (Adriamycin) Hodgkin lymphoma
CHOP cyclophosphamide, hydroxydaunorubicin (doxorubicin), vincristine (Oncovin), prednisone Non-Hodgkin lymphoma
CHOP-R or R-CHOP CHOP + rituximab B-cell non-Hodgkin lymphoma
Clapped clarithromycin, pomalidomide, dexamethasone Multiple myeloma
CMF cyclophosphamide, methotrexate, fluorouracil (5-FU) Breast cancer
CMV cisplatin, methotrexate, vinblastine Transitional bladder carcinoma
CODOX-M cyclophosphamide, vincristine, doxorubicin, high-dose methotrexate Non-Hodgkin lymphoma
COP or CVP cyclophosphamide, Oncovin or vincristine, prednisone Non-Hodgkin lymphoma in patients with history of cardiovascular disease
COPP cyclophosphamide, Oncovin (vincristine), procarbazine, prednisone Hodgkin lymphoma
CT or TC docetaxel (Taxotere), cyclophosphamide Breast cancer
CTD cyclophosphamide, thalidomide, dexamethasone AL amyloidosis
CVAD and Hyper-CVAD cyclophosphamide, vincristine, doxorubicin (Adriamycin), dexamethasone Aggressive non-Hodgkin lymphoma, lymphoblastic lymphoma, some forms of leukemia
CVE carboplatin, vincristin, etoposide Retinoblastoma
CYBORD cyclophosphamide, bortezomib, dexamethasone Multiple myeloma, AL amyloidosis
DA or DAC daunorubicin x 3 days plus ara-C (cytarabine) x 7 days, a variant of 7+3 regimen Acute myeloid leukemia, excluding acute promyelocytic leukemia
DAT daunorubicin, cytarabine (ara-C), tioguanine Acute myeloid leukemia
DCEP dexamethasone, cyclophosphamide, etoposide, platinum agent Relapsed or refractory multiple myeloma
DHAP dexamethasone (a steroid hormone), cytarabine (ara-C), platinum agent Non-Hodgkin lymphoma
DHAP-R or R-DHAP dexamethasone (a steroid hormone), cytarabine (ara-C), platinum agent plus rituximab Non-Hodgkin lymphoma
DICE dexamethasone, ifosfamide, cisplatin, etoposide (VP-16) Aggressive relapsed lymphomas, progressive neuroblastoma
DT-PACE dexamethasone, thalidomide, platinum agent, doxorubicin (Adriamycin), cyclophosphamide, etoposide Multiple myeloma
EC epirubicin, cyclophosphamide Breast cancer
ECF (MAGIC) epirubicin, cisplatin, fluorouracil (5-FU) Gastric cancer and cancer of the esophagogastric junction (Siewert classification III)
EOX epirubicin, oxaliplatin, capecitabine Esophageal cancer, gastric cancer
EP etoposide, platinum agent Testicular cancer, germ cell tumors
EPOCH etoposide, prednisone, vincristine (Oncovin), cyclophosphamide, and hydroxydaunorubicin Non-Hodgkin lymphoma
EPOCH-R or R-EPOCH etoposide, prednisone, vincristine (Oncovin), cyclophosphamide, and hydroxydaunorubicin plus rituximab B-cell non-Hodgkin lymphoma
ESHAP etoposide, methylprednisolone (a steroid hormone), cytarabine (ara-C), platinum agent Non-Hodgkin lymphoma
ESHAP-R or R-ESHAP etoposide, methylprednisolone (a steroid hormone), cytarabine (ara-C), platinum agent plus rituximab Non-Hodgkin lymphoma
FAM fluorouracil, doxorubicin (Adriamycin), mitomycin Gastric cancer
FAMTX fluorouracil, doxorubicin (Adriamycin), methotrexate Gastric cancer
FCM or FMC fludarabine, cyclophosphamide, mitoxantrone B-cell non-Hodgkin lymphoma
FCM-R or R-FCM or R-FMC or FMC-R fludarabine, cyclophosphamide, mitoxantrone plus rituximab B-cell non-Hodgkin lymphoma
FCR fludarabine, cyclophosphamide, rituximab B-cell non-Hodgkin lymphoma
FM fludarabine, mitoxantrone B-cell non-Hodgkin lymphoma
FM-R or R-FM or RFM or FMR fludarabine, mitoxantrone, and rituximab B-cell non-Hodgkin lymphoma
FEC fluorouracil (5-FU), epirubicin, cyclophosphamide Breast cancer
FEC-T fluorouracil (5-FU), epirubicin, cyclophosphamide together, followed by docetaxel (Taxotere) Breast cancer
FL (also known as Mayo) fluorouracil (5-FU), leucovorin (folinic acid) Colorectal cancer
FLAG fludarabine, cytarabine, G-CSF Relapsed or refractory acute myelogenous leukemia
FLAG-Ida or FLAG-IDA or IDA-FLAG or Ida-FLAG fludarabine, cytarabine, idarubicin, G-CSF Relapsed or refractory acute myelogenous leukemia
FLAG-Mito or FLAG-MITO or Mito-FLAG or MITO-FLAG or FLANG mitoxantrone, fludarabine, cytarabine, G-CSF Relapsed or refractory acute myelogenous leukemia
FLAMSA fludarabine, cytarabine, amsacrine Myelodysplastic syndrome, acute myeloid leukemia
FLAMSA-BU or FLAMSA-Bu fludarabine, cytarabine, amsacrine, busulfan Myelodysplastic syndrome, acute myeloid leukemia
FLAMSA-MEL or FLAMSA-Mel fludarabine, cytarabine, amsacrine, melphalan Myelodysplastic syndrome, acute myeloid leukemia
FLOT fluorouracil (5-FU), leucovorin (folinic acid), oxaliplatin, docetaxel Esophageal cancer, gastric cancer
FOLFIRI fluorouracil (5-FU), leucovorin (folinic acid), irinotecan Colorectal cancer
FOLFIRINOX fluorouracil (5-FU), leucovorin (folinic acid), irinotecan, oxaliplatin Pancreatic cancer
FOLFOX fluorouracil (5-FU), leucovorin (folinic acid), oxaliplatin Colorectal cancer
GC gemcitabine, cisplatin gemcitabine, dexamethasone, and cisplatin
GDP gemcitabine, dexamethasone, cisplatin Non-Hodgkin lymphoma and Hodgkin lymphoma
GemOx or GEMOX gemcitabine, oxaliplatin Non-Hodgkin lymphoma
GVD gemcitabine, vinorelbine, pegylated liposomal doxorubicin Hodgkin lymphoma
GemOx-R or GEMOX-R or R-GemOx or R-GEMOX gemcitabine, oxaliplatin, rituximab Non-Hodgkin lymphoma
IA or IAC idarubicin × 3 days plus ara-C (cytarabine) × 7 days, a variant of classic 7+3 regimen Acute myelogenous leukemia, excluding acute promyelocytic leukemia
ICE ifosfamide, carboplatin, etoposide (VP-16) Aggressive lymphomas, progressive neuroblastoma
ICE-R or R-ICE or RICE ICE + rituximab High-risk progressive or recurrent lymphomas
IFL irinotecan, leucovorin (folinic acid), fluorouracil Colorectal cancer
IVA ifosfamide, vincristine, actinomycin D Rhabdomyosarcoma
IVAC ifosfamide, etoposide and high-dose cytarabine Non-Hodgkin lymphoma
m-BACOD methotrexate, bleomycin, doxorubicin (Adriamycin), cyclophosphamide, vincristine (Oncovin), dexamethasone Non-Hodgkin lymphoma
MACOP-B methotrexate, leucovorin (folinic acid), doxorubicin (Adriamycin), cyclophosphamide, vincristine (Oncovin), prednisone, bleomycin Non-Hodgkin lymphoma
MAID mesna, doxorubicin, ifosfamide, dacarbazine Soft-tissue sarcoma
MINE mesna, ifosfamide, novantrone, etoposide Non-Hodgkin lymphoma and Hodgkin lymphoma in relapse or refractory cases
MINE-R or R-MINE mesna, ifosfamide, novantrone, etoposide plus rituximab Non-Hodgkin lymphoma and Hodgkin lymphoma in relapse or refractory cases
MMM mitomycin, methotrexate, mitoxantrone Breast cancer
MOPP mechlorethamine, vincristine (Oncovin), procarbazine, prednisone Hodgkin lymphoma
MVAC methotrexate, vinblastine, doxorubicin (adriamycin), cisplatin Advanced bladder cancer
MVP mitomycin, vindesine, cisplatin Lung cancer and mesothelioma
NP cisplatin, vinorelbine Non–small-cell lung carcinoma
PACE platinum agent, doxorubicin (Adriamycin), cyclophosphamide, etoposide
PCV Procarbazine, CCNU (lomustine), vincristine Brain tumors
PEB cisplatin, etoposide, bleomycin Non-seminomatous germ cell tumors
PEI cisplatin, etoposide, ifosfamide Small-cell lung carcinoma
platin + taxane cisplatin/carboplatin, paclitaxel/docetaxel Ovarian cancer
POMP 6-mercaptopurine (Purinethol), vincristine (Oncovin), methotrexate, and prednisone Acute adult leukemia
ProMACE-MOPP methotrexate, doxorubicin (Adriamycin), cyclophosphamide, etoposide + MOPP Non-Hodgkin lymphoma
ProMACE-CytaBOM prednisone, doxorubicin (Adriamycin), cyclophosphamide, etoposide, cytarabine, bleomycin, vincristine (Oncovin), methotrexate, leucovorin Non-Hodgkin lymphoma
RdC lenalidomide (Revlimid), dexamethasone, cyclophosphamide AL amyloidosis
R-Benda rituximab + bendamustine Follicular lymphoma and MALT lymphoma
R-DHAP or DHAP-R rituximab + DHAP; that is, rituximab, dexamethasone (a steroid hormone), cytarabine (ara-C), platinum agent Relapsed non-Hodgkin lymphoma and Hodgkin lymphoma
R-FCM or FCM-R rituximab + FCM; that is, rituximab, fludarabine, cyclophosphamide, mitoxantrone B-cell non-Hodgkin lymphoma
R-ICE or ICE-R or RICE rituximab + ICE; that is, rituximab, ifosfamide, carboplatin, etoposide High-risk progressive or recurrent lymphomas
RVD lenalidomide (Revlimid), bortezomib, dexamethasone
Stanford V doxorubicin (Adriamycin), mechlorethamine, bleomycin, vinblastine, vincristine, etoposide, prednisone Hodgkin lymphoma
TAC or ACT docetaxel (Taxotere) or paclitaxel (Taxol), doxorubicin (Adriamycin), cyclophosphamide Breast cancer (“TAC” can also refer to tetracaine-adrenaline-cocaine, used as local anesthetic)
TAD tioguanine, cytarabine (ara-C), daunorubicin Acute myeloid leukemia
TC or CT docetaxel (Taxotere), cyclophosphamide Breast cancer
TCH docetaxel (Taxotere), carboplatin, trastuzumab (Herceptin) Breast cancer with positive HER2/neu receptor
TCHP docetaxel (Taxotere), carboplatin, trastuzumab (Herceptin), pertuzumab (Perjeta) Breast cancer with positive HER2/neu receptor
Thal/Dex thalidomide, dexamethasone Multiple myeloma
TIP paclitaxel (Taxol), ifosfamide, platinum agent cisplatin (Platinol) Testicular cancer, germ cell tumors in salvage therapy
EE-4A vincristine, actinomycin Wilms’ tumor
DD-4A vincristine, actinomycin, doxorubicin (Adriamycin) Wilms’ tumor
VABCD vinblastine, doxorubicin (Adriamycin), bleomycin, lomustine (CeeNU), dacarbazine MOPP refractory Hodgkin lymphoma
VAC vincristine, actinomycin, cyclophosphamide Rhabdomyosarcoma
VAD vincristine, doxorubicin (Adriamycin), dexamethasone Multiple myeloma
VAMP one of 3 combinations of vincristine and others Hodgkin lymphoma, leukemia, multiple myeloma
Regimen I vincristine, doxorubicin (Adriamycin), etoposide, cyclophosphamide Wilms’ tumor
VAPEC-B vincristine, doxorubicin (Adriamycin), prednisone, etoposide, cyclophosphamide, bleomycin Hodgkin lymphoma
VD-PACE bortezomib, dexamethasone plus platinum agent, doxorubicin (Adriamycin), cyclophosphamide, etoposide Multiple myeloma
VIFUP vinorelbine, cisplatin, fluorouracil Locally advanced/metastatic breast cancer
VIP vinblastine, ifosfamide, platinum agent, (etoposide(VP-16) may substitute for vinblastine, making a regimen sometimes referred to as VIP-16) Testicular cancer, germ cell tumors
VTD-PACE bortezomib (Velcade), thalidomide, dexamethasone plus platinum agent, doxorubicin (Adriamycin), cyclophosphamide, etoposide Multiple myeloma

Antitumor antibiotics and topoisomerase inhibitors including anthracyclines

Key Points

  • Several of the antibiotic and plant alkaloids and their synthetic versions exert their antitumor action via several mechanisms, the most widely used and best example being anthracyclines.

  • Anthracyclines, including doxorubicin and epirubicin, cause cancer cell death by inhibition of topoisomerase II, intercalation into the DNA, generation of oxidative stress, and induction of mitochondrial dysfunction.

  • These mechanisms also account for cardiotoxicity, although topoisomerase II beta is the target in cardiomyocytes and topoisomerase II alpha in cancer cells.

  • Traditionally acute cardiotoxicity (myocarditis-like presentation) and chronic cardiotoxicity (cardiomyopathy-like presentation) are differentiated.

  • Recovery of cardiac function can be very protracted.

  • Preventive measures include dexrazoxane, the beta-blockers carvedilol, nebivolol, and bisoprolol, angiotensin-converting enzyme (ACE) inhibitors/angiotensin receptor blockers (ARBs), and statins.

Antitumor antibiotics continue to take an important role in the treatment of malignancies (see Table 54.1 ). The anticancer effects of this class of drugs are broad and heavily directed toward the DNA. They include free radical formation with consequent induction of DNA damage or lipid peroxidation, DNA intercalation into the DNA leading to inhibition of macromolecular biosynthesis, and DNA binding, alkylation, and/or crosslinking or interference with DNA unwinding or DNA strand separation and helicase activity. Topoisomerases are enzymes that facilitate the unwinding DNA that is required for normal replication or transcription. Topoisomerase I (TOP I) inhibition leads to single-stranded breaks in DNA, whereas topoisomerase II (TOP II) inhibition causes double-stranded breaks. Accordingly, a distinction of this diverse group of drugs can be made based on their ability to inhibit topoisomerase, and among topoisomerase inhibitors, one can furthermore differentiate between anthracyclines and nonanthracyclines (see Table 54.2 ).

TABLE 54.2
Antitumor Antibiotics and Topoisomerase Inhibitors Including Anthracyclines
TREATMENTS/DRUGS THERAPEUTIC INDICATION MECHANISMS OF CARDIOTOXICITY RISK FACTORS MANIFESTATIONS OF CARDIOTOXICITY PREVENTION AND RISK REDUCTION STRATEGIES
Antitumor Antibiotics
Mitomycin C
  • Gastric cancer

  • Pancreatic cancer

  • Bladder cancer

Unknown
  • High cumulative dose > 30 mg/m 2 [Mitomycin C]

  • Concomitant with anthracycline or non-anthracycline-based chemotherapy

  • Prior coronary artery disease

  • CHF [Mitomycin C]

  • Pericarditis [Bleomycin]

  • Myocardial ischemia/infarction [Bleomycin]

  • Raynaud’s [Bleomycin]

  • Interstitial pneumonitis/fibrosis/pulmonary hypertension [Bleomycin]

  • Dose adjustment and limiting dose of other chemotherapies

  • Supportive care

  • Termination of treatment in case with pericarditis owing to Bleomycin

Bleomycin
  • Lung cancer

  • Lymphomas

  • Germ cell tumors

  • Squamous cell CA

Topoisomerase Inhibitors
Etoposide
  • Testicular cancer

  • SCLC

  • Hematologic cancer

  • Coronary artery vasospasm

  • Direct injury to the myocardium

  • Induction of an immune response

  • Concurrent chemotherapy (esp., bleomycin, cisplatin, ifosfamide)

  • Preexisting CV disease

  • Myocardial ischemia/infarction

  • Vasospastic angina

  • Unknown

Anthracyclines and Analogs
TREATMENTS/DRUGS (MAX. LIMITED DOSE, mg/m 2 ) THERAPEUTIC INDICATION MECHANISMS OF CARDIOTOXICITY RISK FACTORS MANIFESTATIONS OF CARDIOTOXICITY PREVENTION AND RISK REDUCTION STRATEGIES
Doxorubicin (450–500)
Daunorubicin (400–550)
Epirubicin (800–900)
Idarubicin (60)
Breast cancer Gastric tumor
Leukemias
Lymphomas
Lung cancer
Ovarian tumor
Sarcomas
  • ↑Toxic oxygen free radical

  • ↑Oxidative stress

  • Lipid peroxidation of membrane

  • Replacement fibrous tissue

  • ↓Endogenous antioxidant enzyme

  • Iron accumulations complex

  • Cellular apoptosis, DNA damage

  • Topoisomerase IIb inhibition

  • Concurrent chemotherapy

  • Prior/concurrent chest irradiation

  • High dose per cycle (>50 mg/m 2 )

  • High cumulative dose (≥300 mg/m 2 of Doxorubicin, ≥600 mg/m 2 of Epirubicin)

  • IV bolus

  • Extreme age (Elderly >65, children <18)

  • Woman, pregnancy

  • Pre-existing CV disease (CAD, HT, LV dysfunction)

  • Hematopoietic cell transplantation

  • Acute (Initial to several week after therapy)

  • Arrhythmia (AF, SVT, VT, CHB, Mobitz II)

  • ST-T wave abnormalities

  • Anginal pain

  • LV dysfunction/CHF

  • Pericarditis/myocarditis

  • Myocardial ischemia/infarction

  • Chronic (early within 1 year and late onset beyond 1 year after therapy)

  • Asymptomatic diastolic/systolic dysfunction, overt HF

  • Dilated cardiomyopathy

  • Stroke

  • Limiting lifetime cumulative dose

  • Structural modification (Liposome encapsulated molecule) and IV infusion schedule (>6 hours)

  • CV risk assessment before start chemotherapy

  • Intensive monitoring cardiac function during and after therapy

  • F/U ECG if QRS in limb leads ↓≥30% as associated with cardiomyopathy (Daunorubicin)

  • Adjunctive cardioprotective agent

  • Dexrazoxane (Doxorubicin/epirubicin)

  • Beta blocker (carvedilol, nebivolol)

  • ACE-I /ARB

  • Spironolactone

  • Statins

Anthraquinolones
Mitoxantrone (140 mg/m 2 ) Breast cancer, prostate cancer, NHL, AML
Multiple sclerosis
↑Toxic oxygen-free radical
↑Oxidative stress
  • Concurrent Chemotherapy

  • Prior/concurrent chest irradiation

  • High cumulative dose

  • Pericarditis-myocarditis syndrome

  • CHF, arrhythmia

  • Myocardial ischemia/infarction

  • Limiting lifetime cumulative dose

  • CV risk assessment before start chemotherapy

  • Intensive monitoring cardiac function during and after treatment

ALL , Acute lymphoblastic leukemia; ACE-I , angiotensin-converting-enzyme inhibitor; AF , atrial fibrillation; AML , acute myeloid leukemia; CAD , coronary artery disease; CEL , chronic eosinophilic leukemia; CHB , complete heart block; CHF , congestive heart failure; CML , chronic myelogenous leukemia; CT , computed tomography; CV , cardiovascular; CYP3A4 , cytochrome p450 3a4 ; DVT , deep vein thrombosis; ECG , electrocardiogram; GIST , gastrointestinal stroma tumor; GVHD , graft versus host disease; HF , heart failure; HR , heart rate; HT , hormone therapy; HTN , hypertension; ITP , idiopathic thrombocytopenic purpura; LBBB , left bundle branch block; LV , left ventricular; LVH , left ventricular hypertrophy; NHL , non-Hodgkin lymphoma; NCLC , non–small-cell lung carcinoma; PE , pulmonary embolism; RCC , renal cell carcinoma; ROS , reactive oxygen species; RT , radiation therapy; SCD , sudden cardiac death; SCLC , small-cell lung carcinoma; SPECT , single photon emission computerized tomography; SSS , sick sinus syndrome; SVT , supraventricular tachycardia; SVR , stroke volume ratio; TTP , thrombotic thrombocytopenic purpura; VT , ventricular tachycardia; WBC , white blood cells.

Nontopoisomerase inhibitors

The first antibiotic used as an antitumor agent and approved by the US Food and Drug Administration (FDA) in 1964 as such was actinomycin D , obtained from Streptomyces griseus in the mid 1950s. Actinomycin D inhibits the transcription of genes by interacting with a GC-rich duplex, a single-stranded or hairpin form of DNA, and interfering with the action of RNA polymerase.

Mitomycin C was derived in the late 1950s as the second antitumor antibiotic from Streptomyces caespitosus . In tissues, mitomycin C is activated to an alkylating agent and is therefore covered in the following section.

The third prominent antitumor antibiotic in the nontopoisomerase/nonanthracycline category is bleomycin . It was isolated in 1966 from a strain of Streptomyces verticillus. Bleomycin acts by forming complexes with Fe 2+ while binding to DNA. Oxidation of Fe 2+ subsequently leads to the formation of free radicals, which results in DNA damage. Vascular side effects include flushing and Raynaud’s phenomenon and the most concerning toxicity is pulmonary fibrosis (dose-dependent, commonly seen with cumulative doses greater than 300 units, pulmonary function testing with carbon monoxide diffusing capacity is to be obtained before initiation of therapy and with the onset of any symptoms).

Topoisomerase inhibitors

Among the topoisomerase inhibitory antitumor antibiotics anthracyclines hold a special place for several reasons. They were the first group of cancer drugs that was discovered by a joint effort of a pharmaceutical company (Farmitalia) in collaboration with a research center (Istituto Nazionale dei Tumori in Milan). , This effort started in 1960 on a streptomyces strain, Streptomyces peucetius , found near Castel del Monte (Apulia). The new natural antitumor drug was called daunomycin (later daunorubicin), which showed higher efficacy compared with others antitumor drugs in patients with chronic lymphoproliferative diseases. In 1968, with expansion of the joint effort to include Memorial Sloan-Kettering Cancer Center in New York, a new molecule was extracted from a mutated strain of S. peucetius (obtained by treating the microorganism with N-Nitroso-N-methylurea): Adriamycin (later doxorubicin), which showed better activity against tumors in mouse and a greater therapeutic index. It has become the main anthracycline to be used, at least in the United States, for a variety of malignancies. Epirubicin, on the other hand, is used mainly for breast cancer, especially in Europe and idarubicin in the context of induction therapy for acute myeloid leukemia as part of the well-known 7+3 regimen (7 days cytarabine and 3 days idarubicin). For all of these, a key mode of action is inhibition of topoisomerase II alpha, thereby prevention of the relaxation of supercoiled DNA and inhibition of DNA transcription and replication. Additional mechanisms include free radical formation with consequent induction of DNA damage or lipid peroxidation, DNA binding, and alkylation. These two mechanisms are also at play in the myocardium and account for cardiotoxicity. The same holds true for mitoxantrone, which is chemically related to the anthracyclines, an (amino-) anthraquinone that was synthesized in 1979. Unique is its use for metastatic hormone-refractory prostate cancer and in multiple sclerosis.

Currently, the other topoisomerase II inhibitors used clinically is etoposide . Camptothecin , isolated from the Chinese tree Camptotheca acuminate , was used for cancer treatment long before it was identified as a topoisomerase I inhibitor. Owing to its side effects, camptothecin is no longer used clinically. It was replaced by the more effective and safer derivates irinotecan and topotecan .

Cardiovascular toxicities

Anthracyclines are furthermore historically unique because of their unprecedented risk of cardiotoxicity in terms of potency and frequency. In essence, it is only a matter of dose for cardiomyocytes to succumb to anthracyclines. Clinically, two scenarios are differentiated: acute cardiotoxicity, which is a myocarditis-like presentation, and chronic cardiotoxicity, which is a dilated cardiomyopathy-like presentation. Without testing, not many cases declare themselves acutely, even though injury may be present. The inflammation that can be seen in this setting may be classified best as toxic (reactive) myocariditis. Cardiac function assessment (left ventricular ejection fraction [LVEF]) by imaging correlates poorly with cumulative dose and the histologic injury pattern. Heart failure (HF), on the other hand, does seem to have a dose cutoff and follows a decline in cardiac function. Two main mechanisms have prevailed over the years to explain the cardiotoxicity of anthracyclines. The first is the reactive oxygen species (ROS) or iron and free radical theory. Anthracyclines have an affinity for cardiolipin, which brings them in close proximity to the respiratory chain in the mitochondria. In association with anthracyclines cardiolipin is reduced by nicotinamide adenine dinucleotide plus hydrogen (NADH), drawing an electron away from the mitochondrial respiratory chain and subsequently reducing oxygen to form a superoxide anion radical. Further, the addition of an electron to the quinone moiety of anthracyclines leads to the formation of a semiquinone that reverts to the quinone state by reducing molecular oxygen to superoxide anion and its dismutation product hydrogen peroxide (the so called “redox cycling”). A surge in oxidative stress is generated when these products interact with low-molecular iron (Fenton reaction). Anthracyclines may also directly react with ferric iron (Fe3+) to form free radical and alcohol adducts. Collectively these processes lead to oxidative modification of proteins and lipids as well as genomic and mitochondrial DNA damage. Uncoupling of the electron transport chain with impairment of oxidative phosphorylation and adenosine triphosphate synthesis contributes further to mitochondrial dysfunction and damage. Oxidative stress activates a number of stress response pathways, including mitogen-activated protein kinase or extracellular signal-regulated kinase (MAPK/ERK). The phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathway can take a protective role, activated, for instance by neuregulin-1 via human epidermal growth factor receptor-2 (HER2) signaling. This explains the syngergistic action in terms of cardiotoxicity when anthracyclines are combined with trastuzumab. An alternative or at least additional mechanism for anthracycline cardiotoxicity that emerged in recent years is the inhibition of topoisomerase II beta in cardiomyocytes. Interfering with topoisomerase function anthracyclines may impair the repair of DNA as well, not only induce DNA damage, as described above.

Among the other antitumor antibiotics, mainly vascular toxicities have been seen. Mitomycin has been associated with thrombotic microangiopathy and HF on occasion (doses > 30 mg/m 2 ). Bleomycin can cause abnormal vasoreactivity with Raynaud’s phenomenon and chest pain syndromes as clinical presentations. Bleomycin and cisplatin are the two drugs with the highest risk of drug-induced Raynaud’s phenomenon. Cases of acute ischemic events, including myocardial infarction, have been reported as well.

Patients with cardiovascular risk factors are at higher risk. Pericarditis is uncommon; pneumonitis and pulmonary fibrosis (with pulmonary hypertension) rank among the most concerning side effects (in up to 10% of patients).

Among the topoisomerase II inhibitors, irinotecan can cause vasodilation with hypotension and edema. Cases of thromboembolism have been reported, but it is not one of the cancer drugs that have received consistent and widespread alerts in this regard. Etoposide is mainly known for causing hypotension; cases of myocardial ischemia and infarction have been reported as well.

Prevention and treatment of cardiotoxicity

The outlined mechanisms are important for the prevention and treatment of anthracycline cardiotoxicity. Reduction in the level of oxidative stress is a common denominator among agents that have been shown to be beneficial. These include beta blockers with antioxidant properties, ACE inhibitor and angiotensin receptor blockers (ARBs). , Furthermore, statins have been shown to be cardioprotective, possibly related to their antioxidant effects as well as reactivation of the PI3k/AKT kinase pathway. Dexrazoxane has become known as an iron chelator, but also as a strategy to modulate topoisomerase II beta to yield cardioprotective effects.

Alkylating agents

Key Points

  • Alkylating agents act by adding an alkyl group to the guanine base of the DNA molecule, causing structural damage to the DNA strands.

  • Cardiomyopathy, pericarditis, and arrhythmias are the three main cardiovascular toxicities seen with alkylating agents.

  • The risk of cardiovascular side effects is in part dose-related and also influenced by concomitant cardiotoxic chemotherapy.

  • Older patients (age >50, females, those with known coronary artery disease, hypertension or heart failure (HF), prior or concomitant use of anthracycline, or mediastinal radiation are particularly susceptible to worsening HF and are at higher risk for arrhythmias; these patients should be monitored closely and treated for all cardiovascular risk factors.

  • Emerging evidence suggests a role for acrolein in cyclophosphamide cardiotoxicity pointing toward acrolein inhibitors and scavengers as potential cardioprotective agents.

  • No current guidelines exist for monitoring of cardiotoxicity with alkylating agents, but surveillance with echocardiograms and electrocardiograms is prudent, especially when higher doses are used.

  • Early referral to cardiology/cardio-oncology is key in patients with suspected cardiotoxicity.

The recognition of the antitumor effects of alkylating agents dates back to the second decade of the 20th century and World War I as the vesicant properties of mustard gas were shown to induce suppression of lymphoid and hematologic functions. The less-toxic, but closely related, nitrogen mustards of World War II, were selected for further trials in patients with lymphoma and they demonstrated regression of tumors with relief of symptoms. This led to the development of mechlorenthamine as the first alkylating agent used effectively in the treatment of human cancer. , An alternative route led to the approval of platinum drugs, which were discovered during studies on the influence of an electrical field on cell division, leading to the realization that the antiproliferative phenomenon had nothing to do with electricity, but rather with the release of a heavy metal, platinum, from the electrodes.

Although the alkylating agents react with cells in all phases of the cell cycle, their efficacy and toxicity result from interference with rapidly proliferating tissues (see Table 54.3 ). ,

TABLE 54.3
Alkylating Agents
Modified from Hermann J, ed. Clinical Cardio-oncology. Philadelphia: Elsevier; 2018.
TREATMENTS/DRUGS THERAPEUTIC INDICATION MECHANISMS OF CARDIOTOXICITY RISK FACTORS MANIFESTATIONS OF CARDIOTOXICITY PREVENTION AND RISK REDUCTION STRATEGIES
  • Cyclophosphamide

  • Ifosfamide

  • Leukemias

  • Lymphomas

  • Various solid tumors

  • Endothelial capillary damage

  • High dose (not related to cumulative dose)

  • Elderly

  • Prior abnormal ejection fraction

  • Prior irradiation to chest wall

  • Prior anthracyclines

  • Hemorrhagic myocarditis/pericarditis

  • (common 1st week after high dose cyclophosphamide)

  • Pericardial effusion/tamponade

  • LV dysfunction/CHF

  • LVH

  • Corticosteroid and analgesic with termination of treatment

  • Dose adjustment

  • Busulfan

  • Lymphomas

  • Various solid tumors

  • Myocardial fiber fragmentation

  • High-dose regimen

  • Use for lymphoma

  • Arrhythmias, ST-T wave change

  • CHF

  • Dose adjustment

  • ECG monitoring

  • Platinum drugs

  • Cisplatin

  • Carboplatin

  • Oxaliplatin

  • CML

  • Unknown

  • Unknown

  • Endocardial fibrosis/pulmonary fibrosis

  • HTN, arrhythmias

  • Pericardial effusion

  • Germ cell tumors

  • Ovarian cancer

  • Lymphomas

  • Head/neck tumors

  • Lung cancer

  • Sarcomas

  • Anaphylaxis and hypersensitivity reaction

  • Endothelial injury/apoptosis

  • Arterial thrombosis

  • Vascular fibrosis

  • Electrolyte disturbance

  • (hypokalemia, hypomagnesemia)

  • Elderly

  • Prior mediastinal irradiation

  • Used for metastatic testicular cancer

  • Use with cyclophosphamide

  • Early manifestation

    • Venous and arterial thrombotic events

    • Myocardial ischemia/infarction

    • Raynaud’s phenomenon

    • HTN

  • Late manifestation

    • Myocardial Ischemia/infarction

    • HTN, stroke, LVH

    • Arrhythmia (SVT, bradycardia, LBBB)

    • (Ischemic) cardiomyopathy/CHF

    • Raynaud’s phenomenon

  • Adequate hydration and fluid balance

  • Electrolyte check-up and correction

  • Avoid concomitant renal toxic drug

  • Continuous infusion with infusion monitoring for hypersensitivity reaction

CHF , Congestive heart failure; CML , chronic myelogenous leukemia; ECG , electrocardiogram; HTN , hypertension; LVH , left ventricular hypertrophy.

Mchanism of action (see Fig. 54.1 )

The cytotoxicity of alkylating agents can occur in two ways: conventional alkylation and unconventional methylation. Conventional alkylating agents form an aziridinium ion, which is an electrophilic cyclic ion that induces DNA alkylation either directly or through conversion to a carbonium ion. DNA structural change caused by alkylation blocks replication of new DNA (interstrand crosslink formation disrupts DNA separation) and inhibits transcription to mRNA for protein synthesis, impairing cell function, cell growth, and survival. These effects are amplified when alkylating agents form widespread interstrand crosslinks, inhibiting DNA repair and promoting apoptosis.

FIG 54.1, Alkylating agents mechanism of action. Classic alkylating anticancer agents (nitrogen mustard), undergo an intermolecular cyclization reaction to form the electrophilic aziridinium, which causes an alkylating reaction at the nucleophilic N7 of guanine, either directly or through a carbonium ion. This reaction can induce crosslinking when it occurs at two bases on complementary or identical strands. Alkylation of DNA impairs its function, affecting DNA replication and mRNA transcription for protein synthesis. This eventually leads to cellular apoptosis. Depending on the drug, such alkylation can occur only on one base, or on two bases within the same strand inducing crosslinking, or can occur on two bases from the complementary strands inducing interstrand crosslink. Unconventional or nonclassic alkylating agents (dacarbazine) form a methyldiazonium ion that induces methylation by reacting with O6 in guanine. Methylation occurring on a single strand leads to mismatched base pairing of the transformed guanine with thymine instead of cytosine during replication. This results in DNA truncation by DNA repair enzymes, leading to suppression of cell cycle progression or induction of apoptosis.

These conventional alkylating reactions are generally classified through their kinetic properties as SN1 (nucleophilic substitution, first order) or SN2 (nucleophilic substitution, second order). The first-order kinetics of the SN1 reactions depends only on the concentration of the original alkylating agent (e.g., nitrogen mustards or nitrosoureas), which will decide the rate of formation of the reactive intermediate. The SN2 alkylation reaction ( e.g., alkyl sulfonates) is a bimolecular nucleophilic displacement with second-order kinetics, where the rate depends on the concentration of both the alkylating agent and target nucleophile.

In distinction, nonclassic or unconventional alkylating agents are synthetic inorganic nitrogen compounds that induce methylation of guanine bases, and therefore do not have the DNA crosslinking activity shown for classic alkylating agents.

Nevertheless, the therapeutic and toxic effects of alkylating agents do not correlate directly with the subgroup to which they belong or with their chemical reactivity, because clinically useful agents include drugs with SN1 or SN2, and even some with both characteristics. The differences in their toxicity profiles and antitumor activity are more a consequence of their pharmacokinetics, membrane transport, metabolism and detoxification, lipid solubility, penetration of the central nervous system (CNS), and specific enzymatic reactions capable of repairing alkylation sites on DNA.

Classic alkylating agents

Nitrogen mustards

The nitrogen mustards are the most frequently used alkylating agents and the following have replaced mechlorethamine (the original “nitrogen mustard”): cyclophosphamide, ifosfamide, chlorambucil, and melphalan.

Cyclophosphamide is a prodrug that requires activation in the body to release active alkylating species. The initial activation reaction is carried out by cytochrome P450-mediated microsomal oxidation in the liver to produce 4-hydroxycyclophosphamide, after which it readily changes into its isomer aldophosphamide. , In cancer cells, aldophosphamide is broken down into the cytotoxic phosphoramide mustard and the byproduct acrolein. Phosphoramide mustard causes cytotoxicity by inducing DNA crosslinking between guanine molecules. , Cyclophosphamide was observed to have less toxicity in normal tissues, such as liver, bone marrow, and intestinal epidermal cells, compared with previous alkylating drugs. This is owing to the abundant amount of aldehyde-dehydrogenase in these tissues, converting 4-hydroxycyclophosphamide and aldophosphamide to carboxyphosphamide, which is not cytotoxic, is excreted in the urine and accounts for approximately 80% of an administered dose of cyclophosphamide.

Ifosfamide is a structural isomer of cyclophosphamide, with subtle differences in the chemical properties of its reactive metabolite ifosfamide mustard, owing to the location of the chloroethyl group on the nitrogen ring. This makes it less reactive and hence less cytotoxic. , However, the oxidation of this chloroethyl side chain may produce either chloroataldehyde, which has been implicated in the neurotoxicity, or acrolein, which may contribute to the greater renal and bladder toxicity. Sodium-2-mercaptoethane sulfonate (mesna), can convert acrolein into a nontoxic compound and thereby prevent bladder toxicity (hemorrhagic cystitis). Mesna has been used until today to clinically prevent the side effects of ifosfamide, as approved by the FDA for testicular cancer and sarcoma.

Chlorambucil ’s aromatic ring does not form a cyclic aziridinium ion, and therefore the drug exhibits lower toxicity than aliphatic nitrogen mustards. This low reactivity increases the chemical stability of chlorambucil and allows it to reach and stay in the DNA of target cancer cells for a longer time.

Melphalan was developed by conjugating phenylalanine in place of the methyl group in mechlorethamine so that the drug could be delivered specifically to cancer cells through the L-phenylalanine active transport system. Reduced (bone marrow) toxicity and higher cancer cell specificity are the aspired consequences.

Nitrosoureas

Nitrosoureas decompose to produce alkylating and carbamaylating compounds under physiologic conditions to induce DNA damage and to transform proteins, respectively. Representative drugs include carmustine (BCNU) and lomustine (CCNU). Their high lipophilicity allows them to easily penetrate the blood-brain barrier and to be used in CNS malignancies.

Alkyl sulfonates

Busulfan was one of the earliest alkylating agents. It has greater effect on myeloid cells than lymphoid cells (hence its main use in chronic myelogenous leukemia). It exhibits SN2 alkylation kinetics and shows nucleophilic selectivity for thiol groups, suggesting that it may exert cytotoxicity through protein alkylation rather than through DNA modification. ,

Aziridines

Aziridines are analogs of ring-closed intermediates of nitrogen mustards and are less chemically reactive but therapeutically equally effective. These compounds are known to assault cancer cells by triggering DNA interstrand crosslinking. ,

ThioTEPA acts through two mechanisms. The first is forming crosslinks through serial reactions. The second involves hydrolysis to produce an aziridine group, which then forms DNA monoadducts, separating the DNA strands and inducing apoptosis. ,

Mitomycin C was approved by the FDA as a therapeutic agent for lung cancer and pancreatic cancer; however, owing to toxicities, such as hemolysis, and the introduction of other superior anticancer agents, its clinical use has largely declined. ,

Platinum drugs

Platinum drugs remain of major use in the current treatment regimens of various cancers. Discovered by serendipity as mentioned above, cisplatin became the first platinum drug to be approved in 1978 for the treatment of testicular cancer. A variety of platinum complexes were tested thereafter with the collective knowledge of a structure–activity relationship, which was met by carboplatin and oxaliplatin . These platinum complexes, once aquated/activated, can react with nucleophilic centers on purine bases of the DNA (esp., the N7 positions of guanosine and adenosine residues). The two labile coordination sites on the platinum center permit crosslinking of adjacent guanine bases and, to a lesser extent, across different DNA strands to form interstrand crosslinks.

Nonclassic alkylating agents

Hydrazine

Procarbazine was the first alkylating anticancer agent approved by the FDA among hydrazines. Its anticancer effect is by generating a methyldiazonium ion through spontaneous hydrolysis and inducing DNA methylation, which leads to truncation of chromatin threads and apoptosis.

Triazenes

Dacarbazine acts as a prodrug. It becomes demethylated by cytochrome P450 (CYP3A4) and converted into an activated monomethyl compound, which methylates guanine in DNA, causing cytotoxicity. It has the disadvantage of being extremely unstable in aqueous solution and needs to be directly injected into the blood vessels.

Temozolomide is also a prodrug that undergoes spontaneous activation in solution to produce 5-(3-methyltriazen-1-yl) imidazole-4-carboxamide (MTIC), a triazine derivative. It crosses the blood–brain barrier with concentrations in the CNS approximating 30% of plasma concentrations with antitumor activity against gliomas and melanomas.

Cardiovascular toxicities

Alkylating agents can exert cytotoxic effects on the cardiovascular system, often magnified by the comorbid cardiovascular risk factors of an aging population, concomitant cardiotoxic chemotherapies, and radiation. Cardiotoxicity of these agents may range from asymptomatic pericardial effusion to myopericarditis, HF, and arrhythmias (see Table 54.3 ).

Left ventricular dysfunction

Asymptomatic reductions in LVEF as well as symptomatic HF has been reported with alkylating agents, especially with cyclophosphamide, ifosfamide, and mitomycin C.

Cyclophosphamide can cause structural damage, myocarditis, and HF. This risk is dose-dependent, ranging from 7% to 28% with high-dose regimens and becoming evident within 1 to 10 days after administration. Animal studies have suggested that cyclophosphamide by itself is not directly cardiotoxic but acrolein, one of its metabolites, is 1000 times more toxic than doxorubicin. It has been hypothesized that cyclophosphamide (via production of byproducts like acrolein) causes direct endothelial injury, followed by extravasation of toxic metabolites with cardiomyocyte injury, interstitial hemorrhage, and edema ; intracapillary microemboli may develop as well. Myocardial ischemia caused from coronary vasospasm is another proposed mechanism of cardiotoxicity. Moreover, cyclophosphamide impairs cellular respiration and damages the inner mitochondrial membrane of cardiomyocytes, most likely through the induction of oxidative stress. Fulminant HF is seen most frequently in patients receiving a total dose of cyclophosphamide greater than 200 mg/kg prior to bone marrow transplantation. The clinical course is that of rapid onset of severe HF, which can be fatal within 10 to 14 days. The hearts of such patients are dilated, with patchy transmural hemorrhage and pericardial effusion. The microscopic findings consist of interstitial hemorrhage and edema, myocardial necrosis and vacuolar changes, and specific changes in the intramural small coronary vessels. , Cardiotoxicity could be seen in patients receiving lower doses of cyclophosphamide, if used in combination with other alkylating agents. Age above 50 years, previous adriamycin exposure, and concomitant radiation therapy to the left chest appear to increase the risk of cyclophosphamide cardiotoxicity.

Given that ifosfamide and cyclophosphamide are structurally similar, it is possible that ifosfamide may induce HF through a similar mechanism. However, no histopathologic evidence of hemorrhagic myocarditis, which is the hallmark of cyclophosphamide toxicity, was found in patients treated with ifosfamide. Ifosfamide causes nephrotoxicity, which may delay the elimination of cardiotoxic metabolites and lead to acid-base and electrolyte disturbances and thereby cardiac function impairment and arrhythmias.

Mitomycin C , particularly in combination with anthracyclines, may cause cardiomyopathy, probably owing to enhanced oxidative stress. The risk seems to be related to cumulative drug dose (>30 mg/m 2 ).

Arrhythmias

Systemic use of alkylating agents has also been shown to cause a variety of atrial and ventricular tachyarrhythmias in 8% to 10% of patients within 72 hours of administration. , Marked symptomatic sinus bradycardia has also been described. Alkylating agents may cause arrhythmias owing to direct damage of myocytes or through ischemia induced by coronary vasospasm and/or microthrombosis. Histopathologic studies have demonstrated that hypertrophy, fibrosis, and interstitial edema create a substrate vulnerable to arrhythmias. , However, cyclophosphamide and ifosfamide can cause arrhythmias even in the absence of left ventricular dysfunction. Other factors considered to increase the risk of bradyarrhythmias and atrioventricular block include excess vagal tone from severe nausea and emesis. The nephrotoxic effects of ifosfamide with acid-base and electrolyte disturbances may contribute to arrhythmias. Ifosfamide specifically has been shown to cause ventricular tachycardias, atrial fibrillation, and reentry supraventricular tachycardia resistant to pharmacologic therapy.

Supraventricular tachycardia, especially atrial fibrillation (6.6% to 11%), is common after high-dose melphalan. Increased age (>60 years), higher baseline creatinine, larger left atrium size, and previous cardiac comorbidities are noted risk factors.

Pericarditis and pericardial effusion ( Fig. 54.2 )

Cyclophosphamide can cause pericarditis and pericardial effusion to the point of cardiac tamponade. The most feared complication is a hemorrhagic myocarditis/pericarditis, which occurs more commonly after 1 week of the administration of cyclophosphamide with doses greater than 40 mg/kg/day OR 1.4 g/m 2 /day over a minimum of 2 consecutive days. This grave complication presents with HF symptoms, including new dyspnea at rest, elevated jugular venous pulsations, atypical chest pain, and peripheral edema. Tissue pathology demonstrates specific findings of intramyocardial extravasation of blood, fibrin, and fibrin-platelet microthrombi in capillaries. Fibrin strands in the interstitium is the most specific finding for the diagnosis of hemorrhagic myopericarditis. A decrease in QRS voltage can provide a clue. Pericardiocentesis should only be performed if necessary for tamponade.

FIG 54.2, Postmortem examination in a lymphoma patient with fatal cyclophosphamide cardiotoxicity showing fibrinous pericarditis with spotty pericardial hemorrhage ( A ). Cut surface of the myocardium showed diffusely hemorrhagic muscle and bulging appearance of myocardial tissue ( B ). Histologically, there were interstitial edema and extravasation of erythrocytes without infiltration of inflammatory cells or lymphoma cells ( C and D ). Although foci of contraction band necrosis were observed’ most of the cardiac myocytes were histologically preserved.

Busulphan may occasionally cause pericardial and myocardial fibrosis, usually 4 to 9 years later and after a cumulative drug dose of more than 600 mg. Intravenous busulphan may also induce tachycardia, hypertension, or hypotension and LV dysfunction; these complications are not observed after oral drug administration.

Vascular toxicities

Cisplatin (>carboplatin > oxaliplatin) has been associated with vascular events, primarily of two kinds: vasospasm, especially Raynaud’s phenomenon, and thrombosis, including venous and arterial thrombosis. Coronary artery disease does not seem to be a prerequisite for acute coronary events to develop in patients undergoing platinum-based therapies. Intimal erosion secondary to induction of endothelial apoptosis has been suggested as the underlying mechanism (or thromboembolism from an alternate source). Of note, cisplatin levels can remain detectable over decades, indicating a potential long-term risk. Hypertension and renal toxicity are the other main side effects to be aware of for platinum drugs as it pertains to cardio-oncology.

Prevention and treatment of cardiotoxicity

Because most nitrogen mustards have dose-related cardiotoxicity, avoidance of higher doses of these drugs is a key preventative measure.

The relationship between acute toxicity and development of early and late cardiotoxicity is unclear. Clinical trials are required to determine if blocking acute toxicity (e.g., by the use of an ACE inhibitor or ARB) decreases the risk of subsequently developing later cardiotoxicity. All patients who have already developed asymptomatic LV dysfunction or clinical HF, should be treated in keeping with HF guidelines. Of the beta-blockers, carvedilol may have therapeutic advantages over the others, because it has been shown to possess antioxidant properties. Its prophylactic use with alkylating agents is lacking. ,

Management of arrhythmias should be individualized and decisions on the use of antiarrhythmic drugs or device therapy (implantable or external wearable cardioverter defibrillators) should consider the competing risks of cardiac and cancer-related life expectancy, quality of life, and complications. The diligent monitoring of electrolytes, QT interval on electrocardiogram and the use of QT prolonging agents, acid-base disturbances, and catabolic states are also key besides awareness of the direct arrhythmogenic effects of these drugs.

For complications, such as pericarditis, the use of standard therapy with nonsteroidal anti-inflammatory drugs and colchicine is recommended. Pericardiocentesis or pericardial window surgery should be considered in patients with refractory pericarditis or cardiac tamponade, provided they are not coagulopathic or severely thrombocytopenic.

Hemorrhagic myopericarditis with cyclophosphamide and ifosfamide remains highly fatal and no standard treatment options have proven to reduce mortality. Use of corticosteroids, theophylline, nonselective adenosine antagonist, ascorbic acid, or mechanical circulatory support have been anecdotally reported and must be guided by clinical judgement. No reports were found to demonstrate protection with use of coadministration of mesna.

Emerging preventive strategies

Animal studies have suggested that the cardiotoxic effects of cyclophosphamide are mediated through the toxic reactive aldehyde acrolein, inducing extensive protein modifications and myocardial injury. Myocardial glutathione S-transferase P (GSTP), could prevent cyclophosphamide toxicity by detoxifying acrolein. Although this has only been seen in experimental studies in mice, humans with low cardiac GSTP levels or polymorphic forms of GSTP with low acrolein-metabolizing capacity may be more sensitive to cyclophosphamide toxicity.

Methyl palmitate may be of interest owing to its ability to suppress oxidative stress and to interrupt TLR4/NF-κB signaling pathway with reduction of apoptosis. Similarly, by increasing carboxy-ethylphosphoramide mustard and decreasing acroline concentrations and ROS, N-acetylcysteine also has the potential to prevent cyclophosphamide cardiotoxicity. Other agents that have shown promise as cardioprotectants include curcumine and piperine, oral glutamine, kolaviron, silymarin, blueberry anthocyanins-enriched extracts, green tea extract with hydrochlorothiazide, and many others still under clinical investigation.

Antimetabolites

Key Points

  • Antimetabolites act by mimicking the structure of physiologic metabolites that are required for DNA synthesis or by interfering with their native synthesis; based on the metabolite, antifolates (e.g., methotrexate), antipurines (e.g., cytarabine), and antipyrimidines (e.g., 5-fluorouracil [5-FU]) are distinguished.

  • From a cardiovascular perspective, 5-FU has received most attention with a broad spectrum of manifestations including chest pain, ischemia, ST segment elevation myocardial infarction (STEMI), non-STEMI (NSTEMI) arrhythmias, QT prolongation, HF, and stress cardiomyopathy, most of which has been related to altered vasoreactivity.

  • The incidence of 5-FU cardiovascular toxicity is likely less than 1% overall, but 5% or more in patients with established coronary artery disease, most frequently occurring with first exposure.

  • Dose-reduction, bolus infusion, and vasodilator therapy are recommendable management and prevention approaches.

  • Risk/benefit of rechallenge in patients with prior cardiotoxicity should be carefully entertained.

Antimetabolites act by mimicking the structure of physiologic metabolites that are required for DNA synthesis or by interfering with their native synthesis. They most commonly affect cells in the S phase of the cell cycle (cell-cycle specific) when DNA replication is occurring. Based on the metabolite, antifolates, antipurines, and antipyrimidines are distinguished (see Table 54.4 ).

TABLE 54.4
Antimetabolites
Modified from Herrmann J, ed. Clinical Cardio-Oncology . Philadelphia: Elsevier; 2018.
TREATMENTS/DRUGS THERAPEUTIC INDICATION MECHANISMS OF CARDIOTOXICITY RISK FACTORS MANIFESTATIONS OF CARDIOTOXICITY PREVENTION AND RISK REDUCTION STRATEGIES
  • Fluorouracil (5-FU)

  • Capecitabine

  • Breast cancer

  • Colorectal cancer

  • Pancreatic cancer

  • Endothelial cytotoxicity

  • Vascular vasospasm (coronary)

  • Exaggerated sympathetic stimulation

  • Possible direct cytotoxic effect on cardiomyocytes

  • Underlying CV disease

  • Infusion length and dose both in long-term and short-term

  • Previous treatment with 5-FU [Capecitabine]

  • Concomitant chemotherapy

  • Chest pain/angina (5-FU)

  • Myocardial Ischemia/infarction

  • Myocarditis/pericarditis

  • CHF, even cardiogenic shock

  • Stress-induced cardiomyopathy (Takotsubo) (5-FU)

  • Palpitations, arrhythmias, including ventricular tachycardia/fibrillation

  • Sudden cardiac death

  • Termination of treatment (reversible), retreatment after symptom improvement

  • IV bolus regimen, lower dose regimen

  • Antianginal treatment

  • Prophylactic coronary vasodilator therapy (limited efficacy)

  • Fludarabine

  • Pentostatin

  • Cladribine

  • Cytarabine

  • Methotrexate (MTX)

  • Leukemias

  • Lymphomas

  • Various solid tumors

  • Autoimmune Disease

  • Unknown

  • Unknown

  • Pericarditis (pericardial effusion/ tamponade)[cytarabine]

  • Arrhythmia (bradycardia, supra/ventricular arrhythmia) [MTX]

  • Hypotension/chest pain [fludarabine]

  • Myocardial ischemia/infarction

  • CHF

  • Termination of treatment (reversible), retreatment after symptom improvement

  • Steroid treatment in case of pericarditis from cytarabine

  • Antianginal treatment

  • Prophylactic coronary vasodilator (limited efficacy)

CHF , Congestive heart failure; CV , cardiovascular.

Antifolates

Antifolates were the first class of antimetabolites introduced in 1947 with aminopterin inducing complete remission of acute lymphoblastic leukemia. Amethopterin, better known as methotrexate, followed with conserved efficacy but less toxicity. It binds to and inhibits dihydrofolate reductase (DHFR), leading to inhibition of thymidylate and purine synthesis and, subsequently, to the induction of apoptosis.

Methotrexate was rationally designed nearly 60 years ago to potently block DHFR as an antimetabolite, thereby achieving temporary remissions in childhood acute leukemias. DHFR regenerates the reduced form of folate, a requirement for the biosynthesis of purines and thymidylate, which when missing leads to ineffective DNA synthesis and replication. When given in high doses, methotrexate is able to diffuse directly into cells leading to significant toxicity. Therapy is also toxic to nonmalignant cells, although leucovorin is able to bypass DHFR and rescue these cells. The strategy of high-dose therapy followed by leucovorin rescue is commonly used for the treatment of osteosarcoma and hematologic malignancies, especially when there is involvement of the CNS. As with all antimetabolites, methotrexate can cause myelosuppression and mucositis. It can also lead to renal dysfunction, which can be decreased with the use of urine alkalinization. Transaminitis can also be seen. The new generation antifolate pemetrexed is an antimetabolite with activity against certain lung cancers and mesothelioma. It is thought to affect many targets, including thymidylate synthetase, DHFR, and glycinamide ribonucleotide formyltransferase, which ultimately leads to a decreased production of purines and pyrimidines. Folic acid and vitamin B 12 injections are given in conjunction to reduce side effects.

Antipurines and anti(fluoro-) pyrimidines

Antipurines and anti(fluoro-)pyrimidines are analogs to natural deoxynucleotides, competing for the essential role in DNA synthesis, bases, and ribonucleosides. They can be divided into purine analogs (e.g., fludarabine, cladribine ), pyrimidine analog (e.g., cytarabine (ara-C), gemcitabine ), and the fluoropyrimidines (e.g., 5-FU, capecitabine ). 5-FU is readily incorporated into rapidly dividing malignant cells, especially those found within the gastrointestinal tract, explaining the widespread use of 5-FU in gastrointestinal malignancies. 5-FU has multiple mechanisms of action; its metabolite mediates inhibition of thymidylate synthase and 5-FU can also be incorporated into RNA and DNA. 5-FU is clinically administered with leucovorin to increase its antitumor activity. Capecitabine is a prodrug of 5-FU that is selectively activated in tumors overexpressing the activating enzyme thymidine phosphorylase.

5-Fluorouracil is one of the first and most successful designer chemotherapeutics, synthesized in 1957 by Heidelberger and colleagues, as a pyrimidine analog to slow a tumor’s growth by interfering with DNA synthesis. 5-FU-based chemotherapy is the standard of care for adjuvant treatment of stage II and III colorectal cancer (high-risk node-negative and node-positive disease) with a 20% to 30% reduction in mortality. It is also a front-line agent for potentially curable, as well as metastatic, pancreatic cancer, metastatic esophageal and gastric cancer, early stage breast cancer with residual disease after anthracycline-based therapy, and metastatic breast, head, and neck cancers.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here