scispace - formally typeset
Search or ask a question

Why is chemotherapy bad? 


Best insight from top research papers

Chemotherapy is considered bad due to its potential for causing a range of adverse effects on the body. While chemotherapy drugs are designed to target rapidly dividing cancer cells, they can also harm normal cells that divide quickly, leading to side effects like nausea, vomiting, hair loss, fatigue, and more. Moreover, chemotherapy drugs have the capacity to cause long-term damage to vital organs such as the heart, lungs, liver, and nerves. The complexity of the medication process involving chemotherapy also poses risks, with errors in prescribing, preparing, and administering drugs potentially causing harm to patients. Additionally, there are concerns that chemotherapy may not always be beneficial, as it can potentially worsen the patient's condition by disrupting their vital energy.

Answers from top 4 papers

More filters
Papers (4)Insight
Chemotherapy can be harmful due to high toxicity of cytotoxic drugs, narrow therapeutic index, and common errors like wrong doses or drugs, posing risks to patients during treatment.
OtherDOI
21 Mar 2022
Chemotherapy can be harmful due to its strong nature, causing adverse effects like nausea, fatigue, and long-term damage to various organs, although newer agents aim to reduce these effects.
Chemotherapy can have adverse effects due to its targeting of rapidly dividing cells, which can lead to side effects like hair loss, nausea, and weakened immune system.
Chemotherapy may induce more metastases due to its potential harm on vital energy, as per the Arndt Schultz Law. It can worsen patients' condition by spreading cancer throughout the body.

Related Questions

What are the side effects of a chemotherapy?5 answersChemotherapy can cause various side effects. Common physical side effects include weight loss, hair loss, nausea, vomiting, loss of appetite, insomnia, skin discoloration, headache, and fever. Gastrointestinal adverse reactions, such as mucositis, diarrhea, and constipation, are also common. Other side effects include bone marrow suppression, neutrotoxicity, hepatotoxicity, taste change, fatigue, and chest pain. In the case of hyperthermic intraperitoneal chemotherapy (HIPEC) for gastrointestinal cancers, side effects can include enterocutaneous digestive fistulas, GI tract perforation, neutropenia, postoperative bleeding, ventricular tachycardia, hyperglycemia, hypocalcemia, renal impairment, encapsulating peritoneal sclerosis, scrotal ulceration, and sarcopenia. It is important to integrate patient-reported side effects into clinical practice to improve patients' emotional state, quality of life, and compliance with chemotherapy. Further research is needed to understand the mechanisms underlying chemotherapy-induced side effects and to develop strategies for their management.
What influences the chance that side effects of chemotherapy will occur?5 answersThe chance of experiencing side effects from chemotherapy is influenced by various factors. Cancer cells grow rapidly, and chemotherapy drugs target fast-growing cells, which can include both cancerous and healthy cells. Damage to healthy cells leads to side effects. Additionally, patients' expectations before treatment can also play a role. Research suggests that patients' pretreatment expectancies for side effects like nausea, loss of appetite, and feelings of sadness are related to the occurrence of these side effects post-treatment. Furthermore, certain dietary factors can impact the incidence of gastrointestinal symptoms during chemotherapy. Consumption of specific food products such as oils, chocolate, dairy products, stone fruit, and apple can influence the occurrence of nausea, constipation, and diarrhea. Overall, understanding these factors can help healthcare professionals intervene and reduce the burden of chemotherapy-related side effects.
Why the chemotherapy had side effects?5 answersChemotherapy has side effects because the drugs used in chemotherapy target fast-growing cells, which include both cancer cells and healthy cells. These drugs travel throughout the body and can damage normal, healthy cells that are also fast-growing, leading to side effects. The side effects of chemotherapy can vary from mild to severe and can affect both physical and non-physical conditions. Common physical side effects include nausea, weakness, hair loss, vomiting, loss of appetite, weight loss, insomnia, skin discoloration, headache, and fever. Non-physical side effects can include fear of death, impact on work or household duties, feeling anxious about life, and the need to take care of family members. Chemotherapy drugs can also cause long-term damage to various organs in the body. Overall, the side effects of chemotherapy are a result of the drugs' effects on both cancer cells and healthy cells in the body.
What are the common complications of chemotherapy?5 answersChemotherapy can lead to various complications. The most common complications include oral complications such as oral mucositis, oral candidiasis, viral infections, and xerostomia. Another common complication is neurotoxicity, which can manifest as neuropathy, encephalopathy, seizures, headache, vision changes, cerebellar dysfunction, and spinal cord damage. Chemotherapy can also cause myelosuppression, anaemia, neutropenia, thrombocytopenia, thromboembolic disease, cardiovascular disorders, hair loss, nausea, diarrhea, taste change, and fatigue. Additionally, patients may experience bone marrow suppression, gastrointestinal adverse reactions, hepatotoxicity, and taste change. It is important for patients undergoing chemotherapy to be aware of these potential complications and to receive appropriate prophylaxis and treatment to minimize their impact on quality of life and treatment outcomes.
What are the toxicities of asparaginase?3 answersAsparaginase therapy in the treatment of acute lymphoblastic leukemia (ALL) is associated with a range of toxicities. These toxicities include hypersensitivity, hepatotoxicity, hypertriglyceridemia, thromboembolism, pancreatitis, and osteonecrosis. The toxicities of asparaginase are observed more frequently in adult patients compared to children with ALL. The toxicities can be managed by understanding the risk factors and implementing strategies for prevention and management. Pegylated form of asparaginase (PEG asparaginase) is increasingly being used in the treatment of adult ALL, but it still comes with challenges in terms of toxicities. The toxicities associated with pegaspargase include hypersensitivity/anaphylaxis, thrombosis, encephalopathy, hepatitis, and pancreatitis. Age, overweight, and treatment intensity contribute to the toxicities of pegaspargase. Overall, understanding the toxicities of asparaginase and implementing appropriate management strategies is crucial for safe administration of the drug and optimal treatment outcomes.
What are the effects of chemotherapy on patients with colorectal cancer?5 answersChemotherapy can have various effects on patients with colorectal cancer. Subjective reports of cognitive impairment following chemotherapy are frequent, but objective cognitive impairment has been observed regardless of treatment regimen, suggesting a complex relationship between chemotherapy and cognitive function. In patients with compromised performance status, palliative multiagent chemotherapy may improve tumor symptom control, but it does not improve overall survival and carries a risk of toxicity and treatment-related death. Chemotherapy for colorectal cancer typically involves the use of various molecules that target multiple factors contributing to cancer cell growth. Adjuvant chemotherapy with 5-fluorouracil, with or without oxaliplatin, in patients with localized colorectal cancer may lead to a decline in executive function after 12 months compared to patients who did not receive chemotherapy. Between 30% and 40% of colorectal cancer patients receive adjuvant chemotherapy, but the specific effects of chemotherapy on these patients are not mentioned in the abstract.

See what other people are reading

How the apoptosis of nanoparticles are different from free drug solution?
5 answers
The apoptosis induced by nanoparticles loaded with drugs differs from free drug solutions due to enhanced efficacy and targeted delivery. Studies on various nanoparticles like solid lipid nanoparticles (SLNs), poly(ethyleneimine)-functionalized mesoporous silica particles, and redox-activated self-assembled carrier-free nanoparticleshave shown improved cytotoxic activity, lower IC50 values, and selective apoptosis in cancer cells. These nanoparticles exhibit better drug delivery efficiency, increased cellular uptake, and targeted release of drugs, leading to enhanced apoptosis in cancer cells while minimizing toxicity to normal cells. Additionally, the encapsulation of drugs like tamoxifen, doxorubicin, epoxomicin, and curcumin into nanoparticles has shown promising results in inducing apoptosis through various pathways, making them potential candidates for overcoming drug resistance and improving cancer treatment.
How the apoptosis of erlotininb nanoparticles are different from free drug solution?
5 answers
The apoptosis induction of erlotinib nanoparticles differs significantly from the free drug solution. Studies have shown that erlotinib-loaded poly(e-caprolactone) nanocapsules (NCELB) exhibit higher cytotoxicity and induce apoptosis more effectively in A549 lung cancer cells compared to free erlotinib solution. Similarly, erlotinib-loaded NLCs demonstrated enhanced anticancer activity and higher rates of apoptosis in A549 cells compared to erlotinib-liposomes and free erlotinib. Furthermore, self-assembled silk fibroin nanoparticles loaded with erlotinib displayed lower and extended cytotoxicity, indicating a potentially more controlled and sustained apoptotic effect compared to free drugs. These findings collectively suggest that erlotinib nanoparticles have a superior apoptotic impact on cancer cells compared to the free drug solution, highlighting their potential as effective carriers for targeted cancer treatment.
How does the RGD decoration affect the endocytosis of nanoparticles in vivo?
5 answers
The decoration of nanoparticles with RGD peptides significantly influences their endocytosis in vivo. Research has shown that nanoparticles decorated with cyclic RGD peptides exhibit increased internalization by cells expressing specific integrin receptors, such as αvβ3, compared to those decorated with linear RGD peptides. Moreover, the presence of nonconjugated surface moieties on RGD-decorated nanoparticles enhances their recognition by the immune system, affecting their interaction with immune cells, especially phagocytes. Optimizing the RGD content on nanoparticles can lead to enhanced binding with integrin receptors, inhibiting tumor cell metastasis and increasing nanoparticle accumulation at tumor sites. Additionally, RGD-decorated nanoparticles show preferential and persistent accumulation in tumors, indicating improved tumor targeting capabilities. These findings highlight the crucial role of RGD decoration in modulating nanoparticle endocytosis and targeting efficiency in vivo.
Dose of oxaliplatin for colorectal cancer therapy
5 answers
Oxaliplatin is utilized in colorectal cancer therapy at varying doses with different outcomes. Studies have explored different doses and their effects on patients. One study established that a dose of 120 mg/m2 of oxaliplatin in PIPAC procedures showed improved oncological outcomes and quality of life for patients. Another study focused on the combination of statins with oxaliplatin to enhance therapeutic efficacy and reduce neuropathy in KRAS-mutated colorectal cancer, indicating the potential benefits of this combination therapy. Additionally, a case report highlighted dose-dependent reduction in neuropathy symptoms with decreasing oxaliplatin doses, emphasizing the importance of dose titration to manage side effects. Furthermore, the use of oxaliplatin-loaded iodine nanoparticles demonstrated promising results in vitro as a potential chemoradiotherapy agent for colorectal cancer treatment. Overall, these studies provide insights into the diverse approaches and doses of oxaliplatin in colorectal cancer therapy.
Dose of Methotrexate for treatment of colorectal cancer
5 answers
The dose of methotrexate (MTX) for the treatment of colorectal cancer (CRC) can vary depending on the specific context. High-dose methotrexate (HDMTX) is commonly defined as a dose higher than 500 mg/m2 and is utilized in the treatment of various cancers, including CRC. In the study focusing on CRC cells, the enhanced antitumor effect of MTX was investigated in combination with lactate calcium salt (CaLa), which disrupted betaine homeostasis and inhibited methionine synthesis, ultimately enhancing the effectiveness of CRC treatment. Additionally, the administration of high doses of methotrexate in cancer therapy, followed by leucovorin rescue, is crucial to minimize toxicity and prevent serious adverse events. Therefore, for CRC treatment, the dose of methotrexate may be tailored based on individual patient factors and the specific treatment protocol in use.
Why i choose a patient identification between national patient safety goals?
5 answers
Patient identification is a critical aspect of ensuring patient safety in healthcare settings. Proper patient identification processes help in reducing nursing errors and adverse events, ultimately leading to improved quality of care. National Patient Safety Goals emphasize the importance of accurate patient identification to mitigate risks and enhance patient safety. Studies highlight that patient identification significantly impacts the quality of hospital services and patient safety. By focusing on patient identification as part of the National Patient Safety Goals, healthcare providers can ensure that patients receive the right care tailored to their needs, thereby minimizing errors and improving overall healthcare outcomes.
How glutathione impact in child cancer?
5 answers
Glutathione (GSH) plays a significant role in pediatric cancer by influencing disease susceptibility, treatment outcomes, and therapy opportunities. In cancer cells, GSH levels can impact mutagenic mechanisms, DNA synthesis, growth, and resistance to chemotherapy and radiation. While GSH is crucial for detoxification and antioxidation, elevated levels in tumor cells can confer resistance to chemotherapeutic drugs, protecting cells in various types of cancers like bone marrow, breast, colon, larynx, and lung cancers. Studies have highlighted the importance of GSH in promoting cancer progression and impeding chemotherapy, emphasizing the potential of GSH modulation to enhance anti-neoplastic therapy. Understanding the molecular pathways driven by GSH in pediatric cancer could be crucial for developing new therapeutic strategies to combat cancer progression, overcome chemoresistance, and minimize negative side effects.
Are people who take supplements more likely to adhere in research trials?
5 answers
Individuals who take supplements may exhibit varying levels of adherence in research trials. Research suggests that cognitive dysfunction can impact adherence to study protocols, potentially leading to distorted outcomes. Moreover, intentions to use sport supplements can influence responses to interventions, with those intending to use supplements more likely to respond positively. Additionally, older individuals who take more medications, including supplements, may be more likely to use pillboxes for medication management, indicating a potential link between supplement use and adherence behaviors. However, despite the widespread use of vitamin supplements, many trials fail to report results promptly, highlighting issues with compliance and transparency in the supplement research field. Understanding these dynamics is crucial for designing effective research protocols and interventions.
What is the role of IL-34 in the development and progression of cancer?
5 answers
IL-34 plays a significant role in cancer development and progression by influencing the tumor microenvironment and immune responses. It has been found to contribute to chemoresistance by modulating tumor-associated macrophages, promoting immunosuppression, and enhancing cancer cell survival. IL-34 can lead to adverse phenotypes in acute myeloid leukemia (AML) by accelerating disease progression, increasing leukemia stem cell levels, and promoting subcutaneous infiltration of AML cells. Additionally, IL-34 has been associated with resistance to immune checkpoint inhibitors (ICIs) in cancer treatment, suggesting that inhibiting IL-34 activity could potentially overcome ICI resistance and improve therapeutic outcomes. Furthermore, high IL-34 expression has been linked to poor prognosis in ovarian cancer, indicating its role in creating an immunosuppressive tumor microenvironment and conferring resistance to PARP inhibitor therapy.
Impact of infant illness on development and growth?
5 answers
Infant illness, particularly respiratory and febrile illnesses, has a significant negative impact on neurodevelopment during infancy, as highlighted in the research studies conducted on Guatemalan infants. These illnesses are associated with lower Mullen Scales of Early Learning (MSEL) scores at 12-15 months of age, indicating potential developmental delays. However, there was no direct association found between illness and stunting or microcephaly at the same age. The inflammatory response and stress induced by illnesses like acute respiratory infections and fevers can lead to a catabolic state, affecting nutrient handling and overall growth. Proper medical care and attention to infants' health are crucial to mitigate the long-term consequences of these illnesses on growth, development, and neurodevelopmental outcomes.
How effective are bi-specifics in treating multiple myeloma?
5 answers
Bispecific antibodies (BsAbs) have shown significant effectiveness in treating multiple myeloma (MM). BsAbs targeting B-cell maturation antigen (BCMA), GPRC5D, or FcRH5 have demonstrated overall response rates of approximately 60%-70%, with a high proportion of patients achieving very good partial or complete responses. These therapies activate the patient's T-cells to target tumor cells, showing promising results in relapsed/refractory MM patients. BsAbs have been found to induce dose-dependent cell killing of myeloma cells both in vitro and in vivo, leading to tumor regression in xenograft models. Despite their efficacy, BsAbs can lead to adverse events like cytokine release syndrome and infections, emphasizing the importance of monitoring and infectious prophylaxis during treatment. Overall, BsAbs offer a novel and potent therapeutic option for MM patients, with ongoing studies aiming to further enhance their efficacy and safety profiles.