Open access peer-reviewed chapter

Anticancer Activity of Uncommon Medicinal Plants from the Republic of Suriname: Traditional Claims, Preclinical Findings, and Potential Clinical Applicability against Cancer

Written By

Dennis R.A. Mans and Euridice R. Irving

Submitted: 06 August 2018 Reviewed: 27 October 2018 Published: 26 November 2018

DOI: 10.5772/intechopen.82280

From the Edited Volume

Pharmacognosy - Medicinal Plants

Edited by Shagufta Perveen and Areej Al-Taweel

Chapter metrics overview

2,591 Chapter Downloads

View Full Metrics

Abstract

Despite much progress in our understanding of the essence of cancer, remarkable advances in methods for early diagnosis, the expanding array of antineoplastic drugs and treatment modalities, as well as important refinements in their use, this disease is among the leading causes of morbidity and mortality in many parts of the world. In fact, the next decade is anticipated to bring over 20 million new cases per year globally, about half of whom will die from their disease. This indicates a need for better strategies to deal with cancer. One way to go forward is to draw lessons from ancient ethnopharmacological wisdom and to evaluate the plant biodiversity for compounds with potential antineoplastic activity. This approach has already yielded many breakthrough cytotoxic drugs such as vincristine, etoposide, paclitaxel, and irinotecan. The Republic of Suriname (South America), renowned for its pristine and highly biodiverse rain forests as well as its ethnic, cultural, and ethnopharmacological diversity, could also contribute to these developments. This chapter addresses the cancer problem throughout the world and in Suriname, extensively deals with nine plants used for treating cancer in the country, and concludes with their prospects in anticancer drug discovery and development programs.

Keywords

  • cancer
  • Suriname
  • medicinal plants
  • traditional uses
  • phytochemistry
  • pharmacology
  • anticancer activity

1. Introduction

1.1 Generalities

Cancer is a generic term to describe over 200 distinct disease forms that, nonetheless, share three distinguishing characteristics, namely uncontrolled cellular proliferation, invasion of the abnormal cells into adjacent tissues, and their spread to distant organs via blood and lymph vessels [1]. The biological events fundamental to the development of cancer involve the transformation of normal cells to a precancerous lesion which subsequently progresses to a malignant tumor in a multistage process [1]. These changes are the result of the interaction between an individual’s genetic make-up and external agents including physical, chemical, and biological carcinogens [2].

Recognized physical carcinogens are ultraviolet and ionizing radiation which have been linked to skin cancer as well as leukemia and a number of solid tumors, respectively [2]. Well-studied chemical carcinogens are asbestos that has mainly been associated with lung cancer and mesothelioma; components of tobacco smoke which have been linked not only to breast and lung cancer but also to a host of other malignancies; aflatoxins produced by certain molds in improperly stored staple commodities which have been related to liver cancer; and the drinking water contaminant arsenic that has particularly been associated with lung, bladder, and kidney cancer [2]. Examples of biological carcinogens are the human papillomavirus, the hepatitis B virus, the hepatitis C virus, and the Epstein-Barr virus, the causative factors of cervical cancer, liver cancer, and certain lymphomas, respectively; the stomach bacterium Helicobacter pylori that has been implicated in the development of stomach cancer; and certain fish-parasitic flatworms associated with cholangiocarcinoma and urinary bladder cancer [2].

Molecular insights have revealed that the development of cancer—including its capacity to proliferate in an uncontrolled fashion, escape apoptosis, invade neighboring tissues, and disseminate to distant organs—involves aberrations in molecular networks that include oncogenes, tumor suppressor genes, and repair genes [1]. These changes occur in a multistep manner and often take place over many years [1]. This is an important reason that cancer usually manifests at older age, when sufficient carcinogenic mutations have accumulated to cause cancer and innate defense and cellular repair mechanisms have become less effective [1].

1.2 Worldwide epidemiology

According to GLOBOCAN 2018 estimates of cancer incidence and mortality produced by the International Agency for Research on Cancer, cancer will represent the leading cause of death throughout the world in the twenty-first century [3]. In 2018, there were an estimated 18.1 million new cancer cases and 9.6 million cancer deaths globally [3]. Lung cancer and female breast cancer were the most commonly diagnosed malignancy (each 11.6% of total overall cases), followed by cancer of colon and rectum (10.2%), prostate (7.1%), stomach (5.7%), and liver (4.7%) [3]. The most deadly cancers in that year were lung, colorectal, stomach, liver, and breast cancer accounting for 18.4, 9.2, 8.2, 8.2, and 6.6%, respectively, of the total number of cancer fatalities [3]. The most frequent cancers in males were lung, prostate, colorectal, stomach, and liver cancer with incidence rates of 14.5, 13.5, 10.9, 7.2, and 6.3%, respectively, and mortality rates of 22.0, 6.7, 9.0, 9.5, and 10.2%, respectively [3]. And in females, the most common cancers were those of the breast, colon and rectum, lung, and cervix uteri, with incidence rates of 24.2, 9.5, 8.4, and 6.6%, respectively, and mortality rates of 15.0, 9.5, 13.8, and 7.5%, respectively [3].

There were substantial variations among countries with respect to the most frequently diagnosed cancers and the leading causes of cancer death [3]. For instance, for many cancers, incidence rates were generally two- to threefold higher in industrialized countries than in transitioning economies [3]. However, differences in mortality were smaller, as relatively more patients in developing countries died from their disease, probably because of low screening rates as well as less advanced screening services and diagnostic methods in these regions [3]. Furthermore, cancers related to a westernized lifestyle such as lung, breast, and colorectal cancer were (much) more common in industrialized regions than in developing/transitioning regions, even though these neoplasms were among the most common malignancies in both regions [3]. On the other hand, oral cancer and cervical cancer were much more frequent in (certain) developing/transitioning countries than in industrialized countries [3]. These differences are probably for an important part attributable to differences in associated risk factors and screening facilities, respectively, resulting in the former malignancy accounting for almost 50% to the burden of cancer in south-central Asia [4] and the latter occurring at incidence rates between 13.0 and 43.1 per 100,000 in Central America, South America, and the Caribbean, as well as in the parts of Africa [5].

1.3 Treatment modalities

The treatment modalities for cancer depend on the type of cancer as well as its stage and grade [6]. Some cases require only one form of treatment, but most patients need a combination of therapeutic modalities such as surgery with chemotherapy and/or radiation therapy. Surgery is applied for removing localized solid tumors or debulking large solid tumors in order to improve the efficacy of, for instance, chemotherapy [6]. Radiation therapy—external beam radiation therapy, brachytherapy, or systemic radioisotope therapy—uses high doses of radiation to kill cancer cells by damaging their DNA [6]. Chemotherapy is a systemic treatment with mostly combinations of antineoplastic drugs and is intended to kill cancer cells by stopping or slowing their growth or division, but it is also applied as an adjuvant to prevent disease recurrence after surgery or radiation therapy and as a neoadjuvant therapy to decrease the size of a tumor before surgery or radiation therapy [6].

Other cancer treatment modalities are immunotherapy, hormonal therapy, and angiosuppressive therapy. Immunotherapy can make use of adoptive cell transfer involving the infusion of engineered autologous or allogeneic T cells into a patient which can attack the cancer directly; monoclonal antibodies directed at cancer cell-specific antigens; or immunomodulating substances such as cytokines and Bacillus Calmette-Guérin vaccine which stimulate the immune system in a more general way [6]. Hormonal therapy slows or stops the growth of hormone-dependent tumors such as breast and prostate cancers, or reduces or prevents the symptoms in patients suffering from these cancers who do not qualify for surgery or radiation therapy [6]. Hormonal therapy can also be used in the adjuvant or neoadjuvant setting [6]. Angiosuppressive or antiangiogenic therapy interrupts the angiogenic signals that a tumor emits to its surroundings for recruiting a blood supply and causes tumors to shrink [6].

Despite this respectable array of antineoplastic agents and therapeutic modalities most cancers remain fatal, particularly when detected at an advanced stage. This implies a need for more efficacious forms of treatment of neoplastic disease. Many efforts are being dedicated to this goal, including improved early diagnosis, the development of highly specific targeted therapies, and the identification of more efficacious antineoplastic drugs. It is generally agreed that the application of ancient wisdom and folk medicine represents an important strategy to discover and develop new anticancer drugs [7, 8, 9, 10]. This approach has led to breakthrough anticancer drugs such as the tubulin-interfering agents vincristine from the periwinkle plant Catharanthus roseus (L.) G. Don (Apocynaceae) [11] and paclitaxel from the Pacific yew Taxus brevifolia Peattie 1950 (Taxaceae) [12]; the topoisomerase I and II inhibitors irinotecan [13] and etoposide [14], respectively, from Podophyllum plant species (Berberidaceae) and the Chinese happy tree Camptotheca acuminata Decne. (Nyssaceae), respectively; as well as a host of other plant-derived compounds [7, 10]. Notably, almost half of the anticancer drugs that have been granted approval in the United States of America between 1981 and 2014 were from natural origin [9].

So far, only a relative handful of the plant kingdom has been evaluated for pharmacologically active plant substances with potential efficacy against cancer. Therefore, it is likely that further exploration of the rain forests along with other less explored environments such as deserts, tundras, as well as freshwater and marine ecosystems [15], will help identify many structurally novel and mechanistically unique compounds for fighting cancer. This chapter first reviews a few aspects of cancer throughout the world, then focuses on cancer in the Republic of Suriname, subsequently addresses in detail nine medicinal plants that are used for treating cancer in the country, and concludes with some remarks about their potential usefulness against this disease.

Advertisement

2. Background on Suriname

2.1 Geography, population, demographics, and economy

The Republic of Suriname is situated in the north-eastern part of South America adjacent to the Atlantic Ocean and has a land area of roughly 165,000 km2 (Figure 1). The population of about 570,000 is among the ethnically most varied in the world, comprising Amerindians, the original inhabitants; Maroons, the immediate descendants of enslaved Africans shipped from western Africa between the seventeenth and the nineteenth century; Creoles, a generic term referring to anyone having one or more African ancestors; the descendants from indentured laborers attracted from China, India, and Java (Indonesia) between the second half of the nineteenth century and the first half of the twentieth century; as well as immigrants from various European, South American, and Caribbean countries [16].

Figure 1.

Location of Suriname with respect to its neighboring countries French Guiana, Brazil, and Guyana, as well as its position in South America (insert) (from: https://goo.gl/images/F77jgS).

Suriname can be characterized as a demographically transitioning country with declining mortality and infertility rates as well as a growing and aging population. These changes are for an important attributable to considerable progress in health care, nutrition, sanitation, and drinking water quality; the eradication of various infectious diseases; as well as improvements in average living and working conditions, education, and income [17, 18]. The result was a decline of the death rate from 24 per 1000 in 1923 to 6 per 1000 in 2011 and the attainment of an average life expectancy of 70 years in 2011 [17].

The country’s most important economic means of support are crude oil drilling as well as gold and bauxite mining [19]. These activities, together with agriculture, fisheries, forestry, and ecotourism, have substantially contributed to Suriname’s gross domestic income (GDI) in 2014 of USD 5.21 billion and the average per capita income in that year of USD 9325 [19]. This positions Suriname on the World Bank’s list of upper-middle income economies [20].

2.2 Health care

Suriname spends about 5.7% of its GDI—which amounted to USD 589 per capita in 2014—to health care [21]. This sum covers the health costs for the economically weakest individuals; insurance for government employees and employees of government-related companies; import and distribution of essential pharmaceuticals; vaccination programs; maternal and child health care; programs to fight parasitic and microbial diseases; dental care for schoolchildren; services for dermatological diseases, sexually transmitted diseases, and HIV/AIDS; as well as a Kidney Dialyses Center and a Blood Bank [21].

Primary health care in Suriname is offered by the government-subsidized Regional Health Services and Medical Mission, as well as approximately 250 general practitioners. The Regional Health Services run 43 community health centers staffed with physicians and nurses, covers the entire coastal area, and offers basic laboratory testing as well as curative and preventive services including cervical cancer screening and dental, prenatal, and obstetric care. The Medical Mission is a nongovernmental organization that provides health services to people living in Suriname’s hinterland. The clinics are staffed with community health workers who are supervised by general practitioners who travel back and forth on a regular basis.

Secondary care is provided by two private and two government-supported hospitals in Paramaribo and one public hospital in the western district of Nickerie. Medical emergencies can turn around-the-clock to the First-aid Stations of the Academic Hospital Paramaribo and the Saint Vincentius Hospital Suriname. The Academic Hospital Paramaribo also functions as training facility for both general practitioners and medical specialists. All hospitals have modern clinical laboratory facilities as well as radiology services at their disposal. There are, in addition, four private clinical laboratories and three private radiology clinics. Diagnostic imaging including computed tomography and magnetic resonance imaging is possible at two private clinics and the Academic Hospital Paramaribo. This hospital also provides tertiary care at a Thorax Center, a Neurology High-Care Unit, a Neonatal Care Unit, and a Radiotherapy Center.

Advertisement

3. Cancer in Suriname

3.1 Epidemiology

As in many other low- and middle-income countries, there is no population-based cancer registry in Suriname. The occurrence of cancer in the country is estimated from data on the histopathologically confirmed cases at the Pathologic Anatomical Laboratory of the Academic Hospital Paramaribo that functions as the country’s cancer-based registry. This institution reported for 2014 a crude incidence rate of 133 per 100,000 population with the most common cancers being breast, colorectal, prostate, and cervical cancer [22]. An earlier publication [23] mentioned an average of 70 per 100,000 population for the period 1980–2000, suggesting an almost twofold increase in the occurrence of cancer in Suriname since the turn of the century.

Cancer mortality in Suriname has been registered since 1958. In the period between 1962 and 1970, the average death rate due to cancer was 60 per 100,000 per year [24]. This figure had risen to approximately 72 in 2011, ranking cancer as the second most common cause of mortality in the country, after cardiovascular diseases [25]. The top five causes of cancer mortality in that year were prostate, lung, rectum-sigmoid, female breast, and cervical cancer [25]. Most of the fatalities in females were attributable to breast and cervical cancer, while prostate cancer was the leading cause of cancer death in males [25].

3.2 Allopathic forms of cancer treatment in Suriname

Suriname has no national guidelines for the screening, diagnosis, and treatment of cancer, and structured screening programs for breast, cervical, and colon cancer are nonexistent. For these reasons, a comprehensive national cancer control plan has been developed [22] that will be executed in the short term by the Ministry of Health.

Still, primary prevention programs such as mandatory vaccination against the hepatitis B virus (since 2011) and the availability of a HPV vaccine for young girls (implemented in 2013) may help reduce the cancer burden in the country. This may also be achieved by early detection services such as screening for cervical and breast cancer, even though these facilities are in general utilized on an ad hoc basis. Cervical cancer screening occurs upon referral and is done at the Lobi Foundation, a nongovernmental organization for reproductive preventive services, using cytology (Pap smear) or visual inspection with acetic acid. Unfortunately, the coverage of this program is below 20% and thus has probably little impact on cervical cancer mortality [26]. Mammography, breast ultrasound, and fine needle aspiration for the assessment of breast lesions are since 2009 possible at two private clinics and two hospitals. Stereotactic (mammogram-guided) breast biopsy has been available since 2018 at the Academic Hospital Paramaribo. Cancer-specific evaluations such as testing for hormone receptors and tumor markers, are carried out at the Pathologic Anatomical Laboratory of this hospital.

Surgery, radiation therapy, and chemotherapy as standard therapeutic modalities for cancer are all available in Suriname. Surgical treatment is offered by all four hospitals in Paramaribo. Radiation therapy has been available since 2012 and is performed by two radiation oncologists. Chemotherapy is delivered by two oncologists and two gynecologic oncologists. If diagnostic or therapeutic services are not available in Suriname, patients can be transferred to health centers abroad provided that they have a good prognosis and are younger than 70 years. More than half of the selected patients are treated in Bogotá, Columbia. All costs are covered by the Surinamese Ministry of Health through the State Health Foundation [21].

3.3 Traditional forms of cancer treatment in Suriname

All ethnic groups in Suriname have preserved their own specific identity including their particular forms of traditional medicine, probably as a means of strengthening the ethnic identity after their relocation to their new homeland [27, 28]. Not surprisingly, the use of various traditional medicinal systems—involving, among others, Indigenous, African, and Chinese traditional medicine, Indian Ayurveda, as well as Indonesian Jawa—is deeply rooted in Suriname [27, 28]. Furthermore, Suriname’s large biodiversity provides ample and readily available raw material that can be processed into ethnopharmacological plant-based preparations [29]. As a result, many diseases including cancer are often treated with such medications instead of, or in conjunction with, allopathic forms of treatment [30] despite the availability of affordable and accessible modern health care throughout the entire country.

This holds true for, for instance, patients who are motivated by aversion of “chemical” drugs with attendant adverse or side effects and those whose philosophy about life is not compatible with the use of allopathic medicine or who have reservations about the viewpoints of allopathic medicine [31]. Others prefer traditional treatments because these modalities would improve conventional therapies and represent gentler means of managing their disease when compared to allopathic medicines [32]. Still other patients, particularly those with advanced disease or cancer that, from a medical standpoint, can no longer be treated, resort to traditional medicines as an ultimate means to improve their situation [33]. And cultural beliefs, traditional values, and certain perceptions of health and disease may entice some people to choose for a familiar traditional therapy rather than a “western” therapy [34, 35].

Advertisement

4. Plants for treating cancer in Suriname

Hereunder, nine plants that are used in Suriname for treating cancer have in detail been assessed for their presumed activity against this disease. The plants have been selected after consulting a number of comprehensive publications describing various aspects of medicinal plants in the country [36, 37, 38, 39, 40, 41, 42, 43]. Several of these plants such as the graviola Annona muricata L. (Annonaceae), Aloe vera (L.) Burm.f. (Asphodelaceae), the bitter melon Momordica charantia L. (Cucurbitaceae), the neem tree Azadirachta indica A.Juss., 1830 (Meliaceae), Moringa oleifera Lam. (Moringaceae), several subspecies and varieties of the black nightshade Solanum nigrum L. (Solanaceae), as well as the noni Morinda citrifolia L. (Rubiaceae) have elaborately been dealt with in the literature. This led us to decide to leave these plants out of the current chapter and address a number of less well-known plants, which prima facie may not qualify for evaluation for their anticancer potential (Table 1).

FamilySpecies (vernacular names in English; Surinamese)Part(s) usedActive constituent(s)References
AnnonaceaeAnnona squamosa L. (sugar-apple; kaner’apra)Oil from seed, pericarp, leaf, stembarkAnnonaceous acetogenins, terpenes/terpenoids, alkaloids[47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67]
AsteraceaeCyanthillium cinereum (L.) H.Rob. (little ironweed; doifiwiwiri)Whole plantSesquiterpene lactones[74, 75, 76, 77, 78, 79]
AsteraceaeEclipta prostrata (L.) L. (false daisy; luwisa wiwiri)Leaf, aerial parts, whole plantTerthiophenes/thiophenes, saponins, triterpenoids, coumestans, flavonoids[84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94]
FabaceaeAbrus precatorius L., 1753 (crab’s eye; kokriki)Seed, leafAbrin, phenolics, flavonoids[100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110]
FabaceaeTephrosia sinapou (Buch.) Chev. (Surinam poison; bumbi)Root, leaf, aerial parts, stemBenzil derivatives, coumestan derivatives, flavones/flavonoids, phenols[118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131]
LoranthaceaePhthirusa stelis (L.) Kuijt (bird vine; pikin fowru doti)Whole plant, stem, leafPeptides, alkynic fatty acids, lectins, triterpenes, glycosides, flavonoids[135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148]
RubiaceaeUncaria guianensis (Aubl.) J.F. Gmel. (cat’s claw; popokainangra)StembarkOxindole alkaloids[157, 158, 159, 160, 161, 162]
SimaroubaceaeQuassia amara L. (bitterwood; kwasibita)Stem, leafQuassinoids, canthin alkaloids[171, 172, 173, 174, 175, 176, 177, 178]
ZingiberaceaeZingiber officinale (ginger; gember, dyindya)RhizomeGingerols, shogaols[187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203]

Table 1.

Plants with anticancer activity addressed in this chapter, parts preferentially used, presumed constituents with anticancer and chemoprotective activity, and references supporting these activities.

4.1 Annonaceae—Annona squamosa L.

The sugar apple A. squamosa (Figure 2) is probably native to the tropical parts of South America and the Caribbean but is now widely cultivated for its flavorful fruit in many other tropical and subtropical regions throughout the world. Unripe fruits as well as seeds and leaves contain toxic alkaloids with effective vermicidal and insecticidal properties [44]. For these reasons, the seed oil is commonly used to treat head lice [44]. A. squamosa preparations are also used against gastrointestinal ailments, urinary tract infections, irregular menstrual flow, and cancer [42, 43, 45]. The therapeutic efficacy of some of these applications may be attributed to acetogenins, terpenes and terpenoids, as well as alkaloids [45, 46].

Figure 2.

The sugar apple Annona squamosa L. (Annonaceae) (from: https://goo.gl/images/Lh5g7Z).

The seed oil as well as the essential oils from the pericarp, the leaves, and the stembark of A. squamosa displayed anticancer activity against a broad range of human cancer cell lines [47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62] as well as H22 hepatoma implanted into laboratory mice [51, 60, 61, 62, 63]. The anticancer effects have particularly been attributed to annonaceous acetogenins in the seed oil [47, 48, 49, 50, 51, 52, 53, 59] as well as annonaceous acetogenins, terpenes, and terpenoids, and alkaloids in pericarp, leaves, and stembark [56, 57, 58, 60, 61, 62]. Interestingly, the acetogenin squamoxinone-D displayed selective cytotoxicity against the (drug-resistant) SMMC 7721/T cell line [59], and annonaceous acetogens were highly active in H22 hepatoma-bearing laboratory mice [64].

The antitumor activities have in some cases been associated with cycle arrest effects and apoptotic events [54, 60, 61, 62] as indicated by the increased caspase-3 activity, the downregulation of antiapoptotic genes, and the fragmentation of the nuclear DNA [54, 60]. The mechanism underlying these events presumably involves the generation of oxidative stress [54]. This supposition is based on the enhanced generation of intracellular reactive oxygen species and the decreased levels of intracellular glutathione species noted in cultured human cells undergoing apoptosis following exposure to A. squamosa seed oil [54, 60].

Notably, leaf and stembark extracts protected Swiss albino mice and Syrian golden hamsters from the mutagenic effects of the alkylating agent cyclophosphamide [65] or the potent laboratory carcinogen 7, 12 dimethylbenz(a) anthracene (DMBA) [66], respectively. Furthermore, aqueous and ethanolic stembark extracts decreased lipid peroxidation and potentiated antioxidant activities in an animal model of oral carcinogenesis [67]. These observations suggest that A. squamosa also possesses chemopreventive properties.

4.2 Asteraceae—Cyanthillium cinereum (L.) H.Rob.

The little ironweed C. cinereum, also known as Vernonia cinerea (L.) Less. (Figure 3), is native to the tropical parts of Africa and Asia but has become naturalized in various other tropical regions including those in South America and the Caribbean. The plant is traditionally used for treating genitourinary disorders, gastrointestinal complaints, and respiratory ailments; to stimulate perspiration in malaria patients; against childhood conditions including bed-wetting; and to fight cancer [42, 43, 68]. C. cinereum seeds yield vernonia oil that contains vernolic acid [69], a natural epoxy fatty acid that may serve as a renewable starting material for manufacturing adhesives, paints, dyes, coatings, composites, and plastics [70].

Figure 3.

The little ironweed Cyanthillium cinereum (L.) H.Rob. (Asteraceae) (from: https://goo.gl/images/Lh5g7Z).

Pharmacological and phytochemical studies have shown a wide range of bioactive compounds such as (a) sesquiterpene lactone(s), which may lend credit to the traditional uses [68]. Two clinical trials found C. cinereum preparations efficacious in smoking cessation [71], while one study reported encouraging results with a herbal C. cinereum-containing preparation in patients with type 2 diabetes mellitus [72]. However, the clinical evidence available at this moment is insufficiently sound to support these applications [73].

Support for anticancer activity of C. cinereum came from the potent cytotoxicity of an extract from the whole plant against various drug-sensitive and multidrug-resistant human tumor cell lines [74, 75, 76]. The whole-plant extract caused the cells to apoptose and sensitized them to common cytotoxic drugs [76]. Furthermore, such an extract as well as the sesquiterpene lactone vernolide A stimulated the activity of cytotoxic T lymphocytes and natural killer cells and enhanced antibody-dependent cellular cytotoxicity and antibody-dependent complement-mediated cytotoxicity in tumor-bearing BALB/c mice by increasing the secretion of interleukin-2 and interferon-γ [77]. This suggests that (this) sesquiterpene lactone may play an important role in the anticancer activity of C. cinereum [68, 78].

Other indications for anticancer activity of C. cinereum preparations were the inhibitory effects of a 70%-methanol whole-plant extract on the in vitro proliferation, invasion, migration, and matrix metalloproteinase activation of B16F-10 murine melanoma cells [79]. The extract also prevented the formation of lung metastases by the B16F-10 cells in C57BL/6 mice, lowered vascular-endothelial growth factor (VEGF) levels in the animals, and substantially increased their life span when compared to untreated controls [79]. Together, these observations raise the possibility that C. cinereum may exert its anticancer activity by boosting the immune system, suppressing angiogenesis, and inhibiting drug transport mechanisms in addition to direct cytotoxicity.

4.3 Asteraceae—Eclipta prostrata (L.) L.

The false daisy E. prostrata, also known as E. alba (L.) Hassk. or E. erecta L. (Figure 4), is probably native to either Asia or the Americas but is now commonly encountered in subtropical and tropical regions throughout the world. It has become an invasive weed in many parts of the tropics, which is particularly due to its ability to grow fast and flower early. The tender leaves and young shoots are consumed as a vegetable but may also serve as a source for the synthesis of titanium dioxide nanoparticles (nano-TiO2) [80]. Nano-TiO2 is widely employed to provide whiteness and opacity to paints, plastics, papers, inks, food colorants, and toothpastes; for the production of cosmetics and skin care products such as sun blocks because of its ability to protect the skin from UV rays while remaining transparent on the skin; and as an additive in antifogging coatings and self-cleaning windows because of its photocatalytic sterilizing properties [81].

Figure 4.

The false daisy Eclipta prostrata (L.) (Asteraceae) (from: https://goo.gl/images/YGw37Z).

E. prostrata is an important herb in Indian traditional medicine and is used for treating a host of conditions such as skin wounds and certain skin disorders; toothache; hair loss and graying hair; gastrointestinal complaints; uterine disorders; microbial infections; as well as cancer [42, 43, 82]. Some of these claims may be attributable to the presence in the plant of various bioactive constituents including coumestans, thiophene derivatives, terthiophenes, flavonoids, as well as triterpenoids and their glycosides such as eclalbasaponins [82, 83].

Converging lines of evidence suggest that E. prostrata preparations and some of their constituents may elicit anticancer activity through multiple mechanisms including direct cytotoxicity, angiosuppression, and chemoprevention. The former possibility is supported by the growth inhibitory effects of crude extracts of the plant in a variety of drug-sensitive and drug-resistant cell lines [84, 85, 86, 87] while causing apoptosis in some cases [87, 88]. Also, an orally administered methanolic leaf extract exerted encouraging anticancer activity against Ehrlich ascites carcinoma in Swiss albino mice [89], and a hydroalcoholic extract reversed multidrug resistance in an animal model of liver cancer induced by diethylnitrosamine and 2-acetylaminofluorene [86]. Furthermore, terthiophenes, thiophenes, saponins, triterpenoids, coumestans, and flavonoids isolated from the aerial parts exhibited cytotoxicity against cultured SKOV3 human ovarian cancer cells [90]; an eclalbasaponin I-containing fraction from the aerial parts and the saponin dasyscyphin-C isolated from the leaves inhibited the in vitro proliferation of SMMC-7721 human hepatocarcinoma and HeLa human cervical carcinoma cells, respectively [85, 91]; and eclalbasaponin II induced cytotoxicity as well as apoptotic and autophagic cell death in human ovarian cancer cell lines [92].

That E. prostrata may also exhibit angiosuppressive activities can be derived from the inhibitory effect of the juice from the whole plant on invasion, migration, and adhesion of a variety of cancer cell types and endothelial cells in the chick chorioallantoic membrane assay [93]. And indications for chemopreventive actions of this plant were provided by the growth inhibitory effect of a coumastan-containing methanolic whole-plant extract in an experimental skin cancer in mice [94]. This presumably occurred by restoring endogenous antioxidant defense mechanisms, enhancing immunosurveillance, silencing cell cycle progression signals, and inducing stable expression of p53 [94].

4.4 Fabaceae—Abrus precatorius L., 1753

The crab’s eye or rosary pea A. precatorius (Figure 5) is a slender, woody, climbing plant that grows twisting around trees, shrubs, and hedges and probably originates from India. However, due to its severely invasive capacity, this plant is now commonly encountered in many tropical and subtropical parts of the world. Its deep roots are very difficult to remove, and its aggressive growth, hard-shelled seeds, and ability to sucker make it very difficult to eradicate and to prevent re-infestation. The brightly red colored seeds are used to make necklaces and other ornaments as well as percussion instruments in various cultures. However, they are very toxic because of their high content of the toxalbumin abrin, and ingestion of a single well-chewed seed can be fatal [95].

Figure 5.

The crab’s eye Abrus precatorius L., 1753 (Fabaceae) (from: https://goo.gl/images/8VobSd).

The sweet-tasting leaves of the plant are used in West Tropical Africa to sweeten foods [96]. These parts of the plant along with the seeds (after denaturing abrin at high temperatures [97]) are also used in various traditional medicinal systems for treating or preventing tetanus, inflammation, snake bites, rabies, and leukoderma; as aphrodisiacs; as oral contraceptives and abortifacients; and for treating cancer [42, 98].

Pharmacological studies with preparations from A. precatorius seeds and leaves revealed that many of their biological activities may be attributable to abrin [98]. This compound consists of a dimer with a B subunit that facilitates its entry into cells by binding to plasma membrane-associated transport proteins, after which the A subunit inactivates the 26S subunit of ribosomes, preventing protein synthesis [99]. One molecule of abrin is able to inactivate up to 1500 ribosomes per second [95], indicating its powerful inhibitory effect on protein synthesis. On the other hand, this mechanistic feature of abrin presents the opportunity of inhibiting the proliferation of cancerous cells which characteristically have a higher metabolic turnover when compared to normal cells.

Indeed, protein-rich extracts or peptide fractions from A. precatorius seeds and ethanol, ethyl acetate, and water extracts from the leaves potently inhibited the proliferation of several tumor cell lines [100, 101, 102, 103, 104] without affecting the growth of normal murine peritoneal macrophages [102]. The cytotoxic effects were accompanied by upregulation of particularly p21 and p53 levels [104] and clear signs of apoptosis occurring through the mitochondrial pathway [101]. The seed preparations also inhibited the growth of several tumor types implanted into laboratory rodents [105, 106, 107, 108]. And direct injection of abrin into a murine Meth-A sarcoma growing in syngeneic BALB/c mice led to regression of the tumor [109]. The anticancer effects might be related to the antioxidant activities of phenolics and flavonoids in the extracts [102, 104].

Importantly, administration of Meth-A tumor cells which had been treated in vitro with abrin, induced strong antitumor immunity of the mice [109]. This suggests that the antitumor effects of abrin were also produced by boosting the immune system. Support for this presumption came from the immunopotentiating and immunostimulatory properties of abrin [107, 110] and the behavior of Abrus agglutinin as a B cell and T cell stimulator [111].

4.5 Fabaceae—Tephrosia sinapou (Bucholz) A.Chev.

The Surinam poison T. sinapou, also known as T. toxicaria (Swartz) Pers. (Figure 6), is native to parts of Central America, the Caribbean, and tropical South America. The plant is mainly known for its high content of the isoflavonoids rotenone and tephrosin in its black roots and seeds, which are used as a fish poison by the Amazon Indigenous peoples [112, 113]. Particularly rotenone is also highly toxic to insects and pests [112, 113]. For this reason, Guyana hinterland peoples use the root sap or the leaf juice externally against head lice [112]. These preparations are also used to ward off evil spirits and for treating eczema, snakebites, syphilis, and gonorrhea, as well as skin ulcers associated with AIDS and cancer [42, 43, 114]. These health benefits have particularly been ascribed to the rotenoids and other flavonoids in roots, leaves, and aerial parts of the plant [115, 116, 117].

Figure 6.

The Suriname poison Tephrosia sinapou (Bucholz) A.Chev. (Fabaceae) (from: https://goo.gl/images/q6BZZN).

Indications for anticancer activity of Tephrosia preparations were provided by the cytotoxic effects of extracts from parts of T. calophylla Bedd., T. persica Boiss., T. purpurea (L.) Pers., T. villosa (L.) Pers., and T. vogelii Hook F. against human carcinoma cell lines and brine shrimp cultures [118, 119, 120, 121, 122, 123, 124]. In some cases, the cytotoxic effects were accompanied by signs of apoptotic cell death [118]. Comparable anticancer effects were produced by benzil and coumestan derivatives from a T. calophylla root extract [125]; flavonoids from parts of T. calophylla, T. pulcherrima (Baker) Gamble, and T. pumila (Lam.) Pers. [126]; phenol- and flavonoid-rich methanolic extracts from the leaves of T. purpurea and the aerial parts of T. apollinea (Delile) DC. [124, 127]; and a prenylated flavone from the aerial parts of T. apollinea [128]. Notably, the high flavonoid content of the aerial parts of T. apollinea has also been associated with potent anti-angiogenic activity in an ex vivo rat aortic ring assay [127].

There are also indications for cancer chemopreventive activity of Tephrosia preparations. Thus, flavonoids from an ethyl acetate-soluble extract of T. sinapou stem selected for potential cancer chemopreventive properties in an in vitro assay for quinone reductase induction, inhibited the formation of preneoplastic lesions induced by DMBA in a mouse mammary organ culture [129]. Furthermore, T. purpurea extracts substantially reduced the formation of skin lesions in Swiss albino mice treated with the potent tumor promoter phorbol 12-myristate 13-acetate (PMA) following treatment with DMBA [130]. The extract also inhibited the development of hepatocellular carcinoma in Wistar rats treated with the carcinogenic and mutagenic compound N-nitrosodiethylamine [131].

4.6 Loranthaceae—Phthirusa stelis (L.) Kuijt

The bird vine P. stelis (Figure 7) is, like many of its relatives in the plant family Loranthaceae (commonly known as mistletoes), a small flowering plant that grows hemiparasitically on the branches of trees and shrubs. It is encountered in various Southern and Middle American countries between Costa Rica and Bolivia where it often constitutes a serious pest on cultivated trees of economic importance such as rubber, orange, cocoa, and bread fruit trees [132]. P. stelis is mostly spread by bird droppings, hence the abovementioned Surinamese vernacular name of “pikin fowru doti” meaning small birds’ excrement.

Figure 7.

The bird vine Phthirusa stelis (L.) Kuijt (Loranthaceae) (from: https://goo.gl/images/J9D1zG).

None of the parts of the plant have edible uses. However, the viscous layer of its fruits has been suggested to represent a potential source of natural rubber [133]. P. stelis preparations are traditionally used to treat oral candidiasis in children; leukorrhea; problems of the female reproductive system; tonsillitis; and skin problems such as scabies [42, 43, 134]. The plant is also used as a chemopreventive substance and by cancer patients for whom no other options are available, presumably because of its hemiparasitic, cancer-like lifestyle, which would signal its usefulness for these purposes [43].

Indications for anticancer activity of P. stelis are scant, being limited to the cytotoxic effects of small polypeptides of 3–5 kDa isolated from dried dichloromethane or ethanol whole-plant extracts in cultured U-937 GTB human histiocytic lymphoma cells [135]. This finding is in line with the identification of (larger) cytotoxic peptides in the Loranthaceae species Helicanthus elastica (Desr.) Dans. [136] and Ligaria cuneifolia (Ruiz & Pav.) van Tiegh. [137]. Other phytochemicals in Loranthaceae species with in vitro anticancer activity are alkynic fatty acids in Scurrula atropurpurea (BL.) Dans. [138]; lectins in Viscum album coloratum Kom. [139]; the triterpene moronic acid in Phoradendron reichenbachianum (Seem.) Oliv. [140]; glycosides in Macrosolen globosus (Roxb.) Tiegh. [141], Loranthus tanakae Franch. & Sav. (Loranthaceae) [142], and Viscum coloratum (Kom.) Nakai [143]; and flavonoids in L. cuneifolia [144].

Crude extracts from stem or leaves of Scurrula oortiana (Korth.) Danser [145], leaves of Dendrophthoe pentandra (L.) Miq. [146], and stem of Elytranthe parasitica (L.) Danser [147] also exerted cytotoxic effects. In addition, the alkynic fatty acids from S. atropurpurea potently inhibited in vitro tumor cell invasion [148], and extracts from the stem or leaves of S. oortiana increased tumor cell sensitivity to TNF-α-mediated lysis [145].

4.7 Rubiaceae—Uncaria guianensis (Aubl.) J.F. Gmel.

The cat’s claw U. guianensis (Figure 8) is indigenous to the Amazonian parts of Paraguay, Brazil, Bolivia, Peru, Ecuador, Colombia, Venezuela, and the Guyanas. Preparations from its stembark and leaves have a long history of traditional medicinal use and are particularly employed for treating osteoarthritis and rheumatoid arthritis [42, 43, 149]. Pharmacological studies with extracts from U. guianensis—and with those from other closely related species, mainly U. tomentosum (Willd. ex Schult.) DC—indeed showed anti-inflammatory activities [150]. These effects have primarily been attributed to pentacyclic oxindole alkaloids [149, 150, 151, 152]. Clinical studies with an U. guianensis stembark extract or a highly purified pentacyclic oxindole alkaloids fraction from U. tomentosum reported some benefits in patients with osteoarthritis of the knee [153, 154, 155]. However, the overall clinical data are insufficient to draw a firm conclusion about the anti-inflammatory efficacy of Uncaria preparations [156].

Figure 8.

The cat’s claw Uncaria guianensis (Aubl.) J.F. Gmel. (Rubiaceae) (from: https://goo.gl/images/7ezTTN).

No studies have been carried out on the anticancer activity of U. guianensis. However, studies with the oxindole alkaloids from U. tomentosa stembark showed notable anticancer activity against human cancer cell lines [157, 158, 159, 160] and a mouse model [159] which was in some cases accompanied by apoptosis [157]. In addition, Uncaria preparations may possess immunomodulatory and chemopreventive properties besides direct cytotoxic activity. The former assumption is supported by the involvement of anti-inflammatory processes rather than cytotoxic events in the antitumor activity of a hydroethanolic U. guianensis stembark extract in 4 T1 mammary tumor-bearing BALB/c mice [161]. The latter supposition stems from the changes in expression patterns of critical proto-oncogenes and tumor suppressor genes in DMBA-treated CBA/Ca mice following administration of Claw of Dragon tea (CoD™ tea), a mixture of the stembarks from U. guianensis, U. tomentosa, and the trumpet-tree Tabebuia avellanedae Lorentz ex Griseb. (Bignoniaceae) [162].

A clinical trial with a dried extract of U. tomentosa stembark reported improved overall quality of life, social functioning, and fatigue in patients with advanced solid tumors, but there were no improvements in biochemical and inflammatory markers or tumor responses [163]. Another trial found a decrease in the occurrence of neutropenia caused by the 5-fluorouracil-doxorubicin-cyclophosphamide combination in patients with breast cancer [164]. However, a third study found no effect of oral tablets containing a dried ethanolic U. tomentosa stembark on the most prevalent adverse events caused by the 5-fluorouracil-oxaliplatin regimen in colorectal cancer patients [165].

4.8 Simaroubaceae—Quassia amara L.

The bitterwood Q. amara (Figure 9) is native to South and Central America but is now also cultivated in various other tropical and subtropical regions throughout the world. In Suriname, the plant has been named “kwasibita” (Kwasi’s bitter) after the freedman Kwasi or Quassi (1692–1787) who was the first to broadly apply the remarkable medicinal properties of the hardwood for treating malaria fevers [28]. The plant contains triterpene quassinoids, secondary metabolites that are among the bitterest in nature [166]. These compounds are almost exclusively encountered in members of the Simaroubaceae and are a taxonomic marker of this plant family [166]. They constitute basic ingredients of Angostura bitters, concentrated alcoholic preparations produced by the House of Angostura in Trinidad and Tobago, which are key ingredients of cocktails such as gin-based drinks.

Figure 9.

The bitterwood Quassia amara L. (Simaroubaceae) (from: https://goo.gl/images/d5ZqEd).

The quassinoids quassin, neoquassin, bruceantin, and simalikalactones D and E have been associated with a host of pharmacological activities including antimalarial, insecticidal, anti-inflammatory, antimicrobial, and antianorectic activities [166, 167, 168]. Other Q. amara phytochemicals with a broad pharmacological spectrum are canthin-6-one alkaloids, which displayed antiviral, antiparasitic, antibacterial, anti-inflammatory, and cytotoxic activities [166, 168, 169]. Notably, a 4%-Quassia cream containing both groups of phytochemicals has been found safe and effective in the management of rosacea [170].

There is ample evidence that Q. amara preparations and some of its constituents also possess anticancer activity. For instance, crude stem or leaf extracts, quassimarin- and/or similikalactone-enriched fractions, partially purified quassinoid-containing fractions, as well as quassimarin, similikalactones, and canthin alkaloids displayed substantial cytotoxicity against human carcinoma cell lines [171, 172, 173, 174] as well as P-388 lymphocytic leukemia inoculated into laboratory mice [171]. Importantly, the quassinoids did not affect the viability of nontumorogenic African green monkey Vero kidney cells [173] and produced anticancer effects at lower concentrations than those required for antimalarial effects [172, 173]. Comparable results were found with quassinoids and/or canthin alkaloids from other members of the Simaroubaceae family [175, 176, 177, 178]. Markedly, the quassinoids and canthin alkaloids also prevented the activation of Epstein-Barr virus early antigen by PMA [175] and inhibited the activity of CYP1A1, a cytochrome P450 isoform with presumed carcinogen-activating properties [179]. These observations suggest that these compounds may also possess chemopreventive properties.

Based on this large body of preclinical data, several Q. amara constituents have undergone clinical evaluation in patients with advanced solid and hematological malignancies. Unfortunately, the results from phase 1 and phase 2 studies with bruceantin—as well as Fructus bruceae oil obtained from the dried ripe fruits of Brucea javanica (L.) Merr.—were uniformly disappointing, showing no meaningful anticancer activity but substantial toxicity [180, 181, 182].

4.9 Zingiberaceae—Zingiber officinale Roscoe

The ginger Z. officinale (Figure 10) is presumably native to the Indian subcontinent and other Southern Asian regions. This plant was probably introduced in Suriname by Javanese indentured laborers around the beginning of the twentieth century [28, 38]. The rhizome is extensively used as a hot and fragrant kitchen spice in many cuisines and to prepare various hot and cold beverages. This part of the plant also has many long-standing traditional uses [43, 183]. The essential oil from the rhizomes is topically applied as an analgesic, while preparations from powdered fresh or dried rhizomes are orally or topically used for treating, among others, respiratory complaints; obesity; microbial infections; vertigo, travel sickness, morning sickness, as well as nausea and vomiting associated with surgery and chemotherapy; and cancer [43, 183].

Figure 10.

The ginger Zingiber officinale Roscoe (Zingiberaceae) (from: https://goo.gl/images/Tr2fgV).

These claims are supported by the pharmacological activities displayed by particularly gingerols (such as zingerone and zingeberol) and shogaols in the rhizomes. Gingerols are the main compounds in the volatile oil of fresh ginger rhizomes and are responsible for their characteristic fragrance [184]. They are thermally labile and easily undergo dehydration reactions to form the corresponding shogaols, which convey the typical pungent taste of dried ginger during cooking [184]. Both gingerols and shogaols exhibited pharmacological activities which supported the traditional uses of Z. officinale [185, 186].

A host of data supports that gingerols and shogaols possess both anticancer and chemopreventive activities. Evidence for the former suggestion came from their inhibitory effects on the proliferation, cell cycle progression, and viability of human carcinoma cell lines [187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197] and tumors implanted into laboratory animal [198, 199]. Suggestions for chemopreventive activities of these compounds came from their inhibitory effects on the development of cancer in animals treated with laboratory carcinogens [199, 200, 201, 202, 203]. Both activities may be mediated by multiple mechanisms including inhibition of invasion through activation of the nuclear receptor peroxisome proliferator-activated receptor γ (PPAR-γ) [197]; downregulation of matrix metalloproteinase 9 transcription [204]; suppression of tumor angiogenesis [191, 194]; deactivation of aberrant cell cycle-regulating elements [189, 200]; and interference with microtubule integrity [190, 196].

All these observations have led to the consideration of Z. officinale preparations for treating cancer as well as cancer-related complications such as chemotherapy-induced nausea and vomiting. So far, however, there is no scientific proof of clinical efficacy against either cancer [205] or nausea and vomiting resulting from chemotherapy or surgery [206].

Advertisement

5. Concluding remarks

The nine plants addressed in this chapter have a long traditional use in Suriname against various conditions including neoplastic disease and indeed showed some evidence of anticancer activity. However, in all cases, the evidence was limited to preclinical models and was not sufficient to support claims of clinical efficacy. However, this does not necessarily mean that these plants and their active constituents should be discarded as failed compounds. Some may constitute useful parts of an integrative medical approach for treating or preventing cancer. Others—including many mentioned in this chapter—may boost the immune system or improve overall health, well-being, and quality of life. And still others may help relieve some of the symptoms of cancer such as fatigue or reduce the side effects of chemotherapy and radiotherapy.

Converging lines of evidence lend support to these suppositions. Firstly, several phenolic compounds such as curcumin from the turmeric Curcuma longa L. (Zingiberaceae) and apigenin from the celery Apium graveolens L. (Apiaceae) may directly or indirectly exert cytotoxic and apoptotic effects by stimulating autophagy [207, 208]. Other plant phenols such as luteolin in celery, thyme, green peppers, and chamomile tea; epigallocatechin-3-gallate in Chinese green tea; and resveratrol in the skin of grapes, blueberries, raspberries, and mulberries have shown promise in the treatment and prevention of cancer [209, 210, 211]. These compounds are able to inactivate molecular signals and transcription pathways essential for cancer cells, scavenge harmful free radicals, and inhibit tumor angiogenesis, respectively [209, 210, 211].

Secondly, mistletoe extracts may alleviate cancer-related fatigue [212]; preparations from the holy basil Ocimum sanctum L. (Lamiaceae) may avert radiation-induced clastogenesis [213]; those based on A. vera may prevent or treat radiation-induced oral mucositis [214]; and the gingerols and shogaols in Z. officinale may reduce the cardiotoxicity of doxorubicin [215]. These compounds may exercise their protective effects through their anti-inflammatory, immune-modulating, free radical-scavenging, antioxidant, and/or metal-chelating properties [212, 213, 214, 215].

Furthermore, recent advances in analytical and computational techniques as well as the introduction of innovative technologies such as predictive computational software may help employ apparently “useless” anticancer compounds in novel ways. For instance, the rejected tubulin-binding maytansines from Maytenus species (Celastraceae) may have found a new use as “warheads” attached to specific antitumor monoclonal antibodies in order to precisely attack tumor tissues while causing little toxicity [216]. And the discarded topoisomerase I inhibitor lapachol from the stembark of the Surinam greenheart Handroanthus serratifolius (Vahl) S.O. Grose—also known as Tabebuia serratifolia (Vahl) G. Nicholson (Bignoniaceae)—is attracting renewed attention following reports that its inhibitory effect on melanoma cell proliferation may involve interference with glycolysis and decreasing ATP levels [217].

Likewise, the therapeutic index of gingerols in the treatment of breast cancer may improve when formulated as a PEGylated nanoliposomal form, allowing for high specificity, improved bioavailability, slow release, and low systemic toxicity [218]. And structural modifications of quassinoids on the basis of, for instance, (quantitative) structure-activity relationships, may produce more potent and less toxic analogues [219, 220]. These and many other examples support continued assessment of the plants and their bioactive compounds dealt with in the current chapter for their usefulness against cancer. If only one of these compounds would reach the clinic, the efforts invested in their evaluation would have been worthwhile.

Advertisement

Conflict of interest

The authors declare that no conflict of interest exists.

References

  1. 1. Idikio HA. Human cancer classification: A systems biology-based model integrating morphology, cancer stem cells, proteomics, and genomics. Journal of Cancer. 2011;2:107-115
  2. 2. Yadav M, Chatterjee P, Tolani S, Kulkarni J, Mulye M, Chauhan N, et al. A Nexus model of cellular transition in cancer. Biological Research. 2018;51:23. DOI: 10.1186/s40659-018-0173-8
  3. 3. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer Journal for Clinicians. 2018;0:1-31
  4. 4. Shield KD, Ferlay J, Jemal A, Sankaranarayanan R, Chaturvedi AK, Bray F, et al. The global incidence of lip, oral cavity, and pharyngeal cancers by subsite in 2012. CA: A Cancer Journal for Clinicians. 2017;67:51-64
  5. 5. Catarino R, Petignat P, Dongui G, Vassilakos P. Cervical cancer screening in developing countries at a crossroad: Emerging technologies and policy choices. World Journal of Clinical Oncology. 2015;6:281-290
  6. 6. Akulapalli Sudhakar A. History of cancer, ancient and modern treatment methods. Journal of Cancer Science and Therapy. 2009;1:i-iv. DOI: 10.4172/1948-5956.100000e2
  7. 7. Cragg GM, Newman DJ. Natural products: A continuing source of novel drug leads. Biochimica et Biophysica Acta. 2013;1830:3670-3695
  8. 8. Mans DRA. From forest to pharmacy: Plant-based traditional medicines as sources for novel therapeutics. Academia Journal of Medicinal Plants. 2013;1:101-110
  9. 9. Newman DJ, Cragg GM. Natural products as sources of new drugs from 1981 to 2014. Journal of Natural Products. 2016;79:629-661
  10. 10. Iqbal J, Abbasi BA, Mahmood T, Kanwal S, Ali B, Shah SA, et al. Plant-derived anticancer agents: A green anticancer approach. Asian Pacific Journal of Tropical Biomedicine. 2017;7:1129-1150
  11. 11. Van Der Heijden R, Jacobs DI, Snoeijer W, Hallard D, Verpoorte R. The Catharanthus alkaloids: Pharmacognosy and biotechnology. Current Medicinal Chemistry. 2004;11:607-628
  12. 12. Kingston DG, Newman DJ. Taxoids: Cancer-fighting compounds from nature. Current Opinion in Drug Discovery & Development. 2007;10:130-144
  13. 13. Legarza K, Yang LX. Novel camptothecin derivatives. In Vivo. 2005;19:283-292
  14. 14. Baldwin EL, Osheroff N. Etoposide, topoisomerase II and cancer. Current Medicinal Chemistry. Anti-Cancer Agents. 2005;5:363-372
  15. 15. Cragg GM, Newman DJ, Weiss RB. Coral reefs, forests, and thermal vents: The worldwide exploration of nature for novel antitumor agents. Seminars in Oncology. 1997;24:156-163
  16. 16. Algemeen Bureau voor de Statistiek/Censuskantoor. Suriname in cijfers 2013/05. Resultaten achtste (8ste) volks- en woningtelling in Suriname (volume 1) (General Bureau of Statistics/Census The Office Suriname in numbers 2013/05. Results of the Eight General Census of Suriname). Demografische en sociale karakteristieken en migratie (Demographic and social characteristics and migration). Paramaribo: Algemeen Bureau voor de Statistiek; 2013
  17. 17. Oehlers GP, Lichtveld MY, Brewster LM, Algoe M, Irving ER. Health life in Suriname (chapter 6). In: Hassankhan MS, Roopnarine L, White C, Mahase R, editors. Legacy of Slavery and Indentured Labour. Historical and Contemporary Issues in Suriname and the Caribbean. New Delhi: Manohar; 2016. pp. 111-150
  18. 18. Eersel MGM, Vreden SGS, van Eer ED, Mans DRA. Fifty years of primary health care in the rainforest: Temporal trends in morbidity and mortality in indigenous Amerindian populations of Suriname. Journal of Global Health. 2018;8:020423. DOI: 10.7189/jogh.08.020403
  19. 19. Algemeen Bureau voor de Statistiek. Suriname in cijfers 303-2014-04 (General Bureau of Statistics Suriname in Numbers 303-2014-04). Basis Indicatoren (Basic Indicators). Paramaribo: Algemeen Bureau voor de Statistiek; 2014
  20. 20. The World Bank Group. Suriname [Internet]. 2018. Available from: https://data.worldbank.org/country/suriname [Accessed: 10-03-2018]
  21. 21. Ministry of Health. Report of the Director of Health 2005-2007. Paramaribo: Ministry of Health Republic of Suriname; 2008
  22. 22. Dams E. Suriname National Cancer Control Plan 2018-2028. Prepared for the Ministry of Health. Paramaribo: Ministry of Health Republic of Suriname; 2017
  23. 23. Mans DRA, Mohamedradja RN, Hoeblal AR, Rampadarath R, Joe SS, Wong J, et al. Cancer incidence in Suriname from 1980 through 2000 a descriptive study. Tumori. 2003;89:368-376
  24. 24. Lamur HE. The demographic evolution of Surinam, 1920-1970. A sociodemographic analysis (chapter III). In: Lamur HE, editor. Verhandelingen van het Koninklijk Instituut voor Taal-, Land- en Volkenkunde 65 (Discourses of the Royal institute for Linguistics, Land Science, and Ethnology 65). The Hague: Martinus Nijhoff; 1973. pp. 96-98
  25. 25. Punwasi W. Causes of death in Suriname 2010-2011. Bureau Openbare Gezondheidsdienst (Bureau of Public Health). Paramaribo: Ministry of Health Republic of Suriname; 2012
  26. 26. ER I, DRA M. Age and ethnic differences in the occurrence of cervical dysplasia, cervical cancer and cervical cancer deaths in Suriname. Translational Biomedicine. 2015;6:1. DOI: 10.21767/2172-0479.100001
  27. 27. Mans DRA. “Nature, green in leaf and stem”. Research on plants with medicinal properties in Suriname. Clinical and Medical Investigations. 2016;2:1-10
  28. 28. Mans DRA, Ganga D, Kartopawiro J. Meeting of the minds: Traditional herbal medicine in multiethnic Suriname (chapter 6). In: El-Shemy H, editor. Aromatic and Medicinal Plants—Back to Nature. Rijeka: InTech; 2017. pp. 111-132. DOI: 10.5772/66509
  29. 29. Hammond DS. Forest conservation and management in the Guiana shield (chapter 1). In: Hammond DS, editor. Tropical Rainforests of the Guiana Shield. Wallingford: CABI Publishing; 2005. pp. 1-14
  30. 30. Yue Q, Gao G, Zou G, Yu H, Zheng X. Natural products as adjunctive treatment for pancreatic cancer: Recent trends and advancements. BioMed Research International. 2017;2017:8412508. DOI: 10.1155/2017/8412508
  31. 31. Marinac JS, Buchinger CL, Godfrey LA, Wooten JM, Sun C, Willsie SK. Herbal products and dietary supplements: A survey of use, attitudes, and knowledge among older adults. The Journal of the American Osteopathic Association. 2007;107:13-23
  32. 32. Sparber A, Bauer L, Curt G, Eisenberg D, Levin T, Parks S, et al. Use of complementary medicine by adult patients participating in cancer clinical trials. Oncology Nursing Forum. 2000;27:623-630
  33. 33. Mansky PJ, Wallerstedt DB. Complementary medicine in palliative care and cancer symptom management. Cancer Journal. 2006;1:425-431
  34. 34. Daher M. Cultural beliefs and values in cancer patients. Annals of Oncology. 2012;23(Suppl 3):66-69
  35. 35. Luo T, Spolverato G, Johnston F, Haider AH, Pawlik TM. Factors that determine cancer treatment choice among minority groups. Journal of Oncology Practice/American Society of Clinical Oncology. 2015;11:259-261
  36. 36. Stephen HJM. Geneeskruiden van Suriname: Hun toepassing in de volksgeneeskunde en in de magie (Herbal Medicines from Suriname: Their Applications in Folk Medicine and Wizardry). Amsterdam: De Driehoek; 1979
  37. 37. Heyde H. Surinaamse medicijnplanten (Surinamese Medicinal Plants). 2nd ed. Paramaribo: Westfort; 1987
  38. 38. Tjong Ayong G. Het gebruik van medicinale planten door de Javaanse bevolkingsgroep in Suriname (The Use of Medicinal Plants by the Javanese in Suriname). Paramaribo: Instituut voor de Opleiding van Leraren; 1989
  39. 39. Slagveer JL. Surinaams Groot Kruidenboek: Sranan Oso Dresie (Surinamese Herbal Medicines). Paramaribo: De West; 1990
  40. 40. Sedoc NO. Afrosurinaamse natuurgeneeswijzen: Bevattende meer dan tweehonderd meest gebruikelijke geneeskrachtige kruiden (Afro-Surinamese Natural Remedies: Over Two Hundred Commonly Used Medicinal Herbs). Paramaribo: Vaco Press; 1992
  41. 41. Raghoenandan UPD. Etnobotanisch onderzoek bij de Hindoestaanse bevolkingsgroep in Suriname (An ethnobotanical investigation among hindustanis in Suriname) [thesis]. Paramaribo: Anton de Kom University of Suriname; 1994
  42. 42. DeFilipps RA, Maina SL, Crepin J. Medicinal Plants of the Guianas (Guyana, Surinam, French Guiana). Washington, DC: Smithsonian Institution; 2004
  43. 43. Van Andel TR, Ruysschaert S. Medicinale en rituele planten van Suriname (Medicinal and Ritual Plants of Suriname). Amsterdam: KIT Publishers; 2011
  44. 44. Zahid M, Mujahid M, Singh PK, Farooqui S, Singh K, Parveen S, et al. Annona squamosa Linn. (custard apple): An aromatic medicinal plant fruit with immense nutraceutical and therapeutic potentials. International Journal of Pharmaceutical Sciences and Research. 2018;9:1745-1759
  45. 45. Saha R. Pharmacognosy and pharmacology of Annona squamosa: A review. International Journal of Pharmacy and Life Sciences. 2011;2:1183-1189
  46. 46. Oo WM, Khine MM. Pharmacological activities of Annona squamosa: Updated review. International Journal of Pharmaceutical Chemistry. 2017;3:86-93
  47. 47. Xie H, Wei J, Liu M, Yang R. A new cytotoxic acetogenin from the seeds of Annona squamosa. Chinese Chemical Letters. 2003;14:588
  48. 48. Liaw CC, Yang YL, Chen M, Chang FR, Chen SL, Wu SH, et al. Mono-tetrahydrofuran annonaceous acetogenins from Annona squamosa as cytotoxic agents and calcium ion chelators. Journal of Natural Products. 2008;71:764-771
  49. 49. Yang HJ, Zhang N, Chen JW, Wang MY. Two new cytotoxic acetogenins from Annona squamosa. Journal of Asian Natural Products Research. 2009;11:250-256
  50. 50. Chen Y, Chen JW, Li X. Cytotoxic bistetrahydrofuran annonaceous acetogenins from the seeds of Annona squamosa. Journal of Natural Products. 2011;74:2477-2481
  51. 51. Chen Y, Xu SS, Chen JW, Wang Y, Xu HQ, Fan NB, et al. Anti-tumor activity of Annona squamosa seeds extract containing annonaceous acetogenin compounds. Journal of Ethnopharmacology. 2012;142:462-466
  52. 52. Chen Y, Chem J, Wang Y, Xu S, Li X. Six cytotoxic annonaceous acetogenins from Annona squamosa seeds. Food Chemistry. 2012;135:960-966
  53. 53. Miao Y, Xu X, Yuan F, Shi Y, Chen Y, Chen J, et al. Four cytotoxic annonaceous acetogenins from the seeds of Annona squamosa. Natural Product Research. 2016;30:1273-1279
  54. 54. Pardhasaradhi BV, Reddy M, Kumari AM, Ali AL, Khar A. Differential cytotoxic effects of Annona squamosa seed extracts on human tumor cell lines: Role of reactive oxygen species and glutathione. Journal of Biosciences. 2005;30:237-244
  55. 55. Nakano D, Ishitsuka K, Kamikawa M, Matsuda M, Tsuchihashi R, Okawa M, et al. Screening of promising chemotherapeutic candidates from plants against human adult T-cell leukemia/lymphoma. Journal of Natural Medicines. 2013;67:894-903
  56. 56. Li XH, Hui YH, Rupprecht JK, Liu YM, Wood KV, Smith DL, et al. Bullatacin, bullatacinone, and squamone, a new bioactive acetogenin, from the bark of Annona squamosa. Journal of Natural Products. 1990;53:81-86
  57. 57. Hopp DC, Alali FQ, Gu ZM, McLaughlin JL. Three new bioactive bis-adjacent THF-ring acetogenins from the bark of Annona squamosa. Bioorganic & Medicinal Chemistry. 1998;6:569-575
  58. 58. Sun L, Zhu H, Gan L, Mo J, Feng F, Zhou C. Constituents from the bark of Annona squamosa and their anti-tumor activity. Zhongguo Zhong Yao Z Zhi. 2012;37:2100-2104
  59. 59. Vilanova NS, Morais SM, Facao MJ, Machado LM, Becilaqua CM, Costa IR, et al. Leishmanicidal activity and cytotoxicity of compounds from two Annonacea species cultivated in Northeastern Brazil. Revista da Sociedade Brasileira de Medicina Tropical. 2011;44:567-571
  60. 60. Ma C, Wang Q, Shi Y, Li Y, Wang X, Li X, et al. Three new anti-tumor annonaceous acetogenins from the seeds of Annona squamosa. Natural Product Research. 2017;31:2085-2090
  61. 61. Jou B, Remanin P. Antitumor constituents from Annona squamosa fruit pulp. Medicinal Chemistry Research. 2008;17:345-355
  62. 62. Chen YY, Cao YZ, Li FQ, Xl Z, Peng CX, Lu JH, et al. Studies on anti-hepatoma activity of Annona squamosa L. pericarp extract. Bioorganic & Medicinal Chemistry Letters. 2017;27:1907-1910
  63. 63. Chen YY, Peng CX, Hu Y, Bu C, Guo SC, Li X, et al. Studies on chemical constituents and anti-hepatoma effects of essential oil from Annona squamosa L. pericarp. Natural Product Research. 2017;31:1308-1308
  64. 64. Chen Y, Shi Y, Ma C, Wang X, Li Y, Miao Y, et al. Antitumor activity of Annona squamosa. Journal of Ethnopharmacology. 2016;193:362-367
  65. 65. Yang RM, Li WM, Hu WJ, Huang WH, Zhu CY, Yu JG, et al. Anticancer effect of total annonaceous acetogenins on hepatocarcinoma. Chinese Journal of Integrative Medicine. 2015;21:682-688
  66. 66. Thakkar JH, Solanki HK, Tripathi P, Patel NJ, Jani GK. Evaluation of antimutagenic potential of Annona squamosa leaf extract. Elixir Human Physiology. 2011;31:1960-1965
  67. 67. Suresh K, Manoharn S, Blessy D. Protective role of Annona squamosa Linn bark extracts in DMBA induced genotoxicity. Kathmandu University Medical Journal. 2008;6:364-369
  68. 68. Suresh K, Manoharan S, Panjamurthy K, Kavitha K. Chemoprotective and antilipidperoxidative efficacy of Annona squamosa bark extracts in experimental oral carcinogenesis. Pakistan Journal of Biological Sciences. 2006;9:2600-2605
  69. 69. Joshi RK. GC/MS analysis of the essential oil of Vernonia cinerea. Natural Product Communications. 2015;10:1319-1320
  70. 70. Jaworski J, Cahoon EB. Industrial oils from transgenic plants. Current Opinion in Plant Biology. 2003;6:178-184
  71. 71. Wongwiwatthananukit S, Benjanakaskul P, Songsak T, Suwanamajo S, Verachai V. Efficacy of Vernonia cinerea for smoking cessation. Journal of Health Research. 2009;23:31-36
  72. 72. Bin Sayeed MS, Mostofa AG, Ferdous FM, Islam MS. A randomized, placebo-controlled, crossover study of an herbal preparation containing Vernonia cinerea in the treatment of type 2 diabetes. Journal of Alternative and Complementary Medicine. 2013;19:767-771
  73. 73. Puttarak P, Pornpanyanukul P, Meetam T, Bunditanukul K, Chaiyakunapruk N. Efficacy and safety of Vernonia cinerea (L.) Less. for smoking cessation: A systematic review and meta-analysis of randomized controlled trials. Complementary Therapies in Medicine. 2018;37:37-42
  74. 74. Khay M, Toeng P, Mahiou-Leddet V, Mabrouki F, Sothea K, Ollivier E, et al. HPLC analysis and cytotoxic activity of Vernonia cinerea. Natural Product Communications. 2012;7:1259-1262
  75. 75. Guha G, Rajkumar V, Ashok Kumar R, Mathew L. Therapeutic potential of polar and non-polar extracts of Cyanthillium cinereum in vitro. Evidence-based Complementary and Alternative Medicine. 2011;2011:784826. DOI: 10.1093/ecam/nep155
  76. 76. Appadath Beeran A, Maliyakkal N, Rao CM, Udupa N. The enriched fraction of Vernonia cinerea L. induces apoptosis and inhibits multi-drug resistance transporters in human epithelial cancer cells. Journal of Ethnopharmacology. 2014;158(Pt A):33-42
  77. 77. Pratheeshkumar P, Kuttan G. Modulation of cytotoxic T lymphocyte, natural killer cell, antibody-dependent cellular cytotoxicity, and antibody-dependent complement-mediated cytotoxicity by Vernonia cinerea L. and vernolide-A in BALB/c mice via enhanced production of cytokines IL-2 and IFN-γ. Immunopharmacology and Immunotoxicology. 2012;34:46-55
  78. 78. Shoaib M, Shah I, Ali N, Adhikari A, Tahir MN, Shah SWA, et al. Sesquiterpene lactone! a promising antioxidant, anticancer and moderate antinociceptive agent from Artemisia macrocephala Jacquem. BMC Complementary and Alternative Medicine. 2017;17:27. DOI: 10.1186/s12906-016-1517-y
  79. 79. Pratheeshkumar P, Kuttan G. Modulation of immune response by Vernonia cinerea L. inhibits the proinflammatory cytokine profile, iNOS, and COX-2 expression in LPS-stimulated macrophages. Immunopharmacology and Immunotoxicology. 2011;33:73-83
  80. 80. Rajakumar G, Abdul Rahuman A, Priyamvada B, Gopiesh Khanna V, Kishore Kumar D, Sujin PJ. Eclipta prostrata leaf aqueous extract mediated synthesis of titanium dioxide nanoparticles. Materials Letters. 2012;68:115-117
  81. 81. Tamimi H, Shishesaz MR, Farzam M, Jafari D. A review on nanoparticles of titanium dioxide: Characteristics, methods of synthesis and their application in organic coatings. International Journal of Advanced Biotechnology and Research. 2016;7:1226-1231
  82. 82. Chung IM, Rajakumar G, Lee JH, Kim SH, Thiruvengadam M. Ethnopharmacological uses, phytochemistry, biological activities, and biotechnological applications of Eclipta prostrata. Applied Microbiology and Biotechnology. 2017;101:5247-5257
  83. 83. Mithun NM, Shashidhara S, Vivek Kumar R. Eclipta alba (L.). A review on its phytochemical and pharmacological profile. Pharmacology. 2011;1:345-357
  84. 84. Lee MK, Ha NR, Yang H, Sung SH, Kim GH, Kim YC. Antiproliferative activity of triterpenoids from Eclipta prostrata on hepatic stellate cells. Phytomedicine. 2008;15:775-780
  85. 85. Liu QM, Zhao HY, Zhong XK, Jiang JG. Eclipta prostrata L. phytochemicals: Isolation, structure elucidation, and their antitumor activity. Food and Chemical Toxicology. 2012;50:4016-4022
  86. 86. Chaudhary H, Dhuna V, Singh J, Kamboj SS, Seshadri S. Evaluation of hydro-alcoholic extract of Eclipta alba for its anticancer potential: An in vitro study. Journal of Ethnopharmacology. 2011;136:363-367
  87. 87. Yadav NK, Arya RK, Dev K, Sharma C, Hossain Z, Meena S, et al. Alcoholic extract of Eclipta alba shows in vitro antioxidant and anticancer activity without exhibiting toxicological effects. Oxidative Medicine and Cellular Longevity. 2017;2017:9094641. DOI: 10.1155/2017/9094641
  88. 88. Chauhan N, Singh D, Painuli RM. Screening of bioprotective properties and phytochemical analysis of various extracts of Eclipta alba whole plant. International Journal of Pharmacy and Pharmaceutical Sciences. 2012;4:554-560
  89. 89. Gupta M, Mazumdera UK, Haldar PK, Kandar CC, Manikanda L, Senthil GP. Anticancer activity of Indigofera aspalathoides and Wedelia calendulaceae in Swiss albino mice. Iranian Journal of Pharmaceutical Research. 2007;6:141-145
  90. 90. Kim HY, Kim HM, Ryu B, Lee JS, Choi JH, Jang DS. Constituents of the aerial parts of Eclipta prostrata and their cytotoxicity on human ovarian cancer cells in vitro. Archives of Pharmacal Research. 2015;38:1963-1969
  91. 91. Khanna VG, Kannabiran K. Anticancer-cytotoxic activity of saponins isolated from the leaves of Gymnema sylvestre and Eclipta alba on HeLa cells. International Journal of Green Pharmacy. 2009;1:227-229
  92. 92. Cho YJ, Woo JH, Lee JS, Jang DS, Lee KT, Choi JH. Eclalbasaponin II induces autophagic and apoptotic cell death in human ovarian cancer cells. Journal of Pharmacological Sciences. 2016;132:6-14
  93. 93. Lirdprapamongkol K, Kramb JP, Chokchaichamnankit D, Srisomsap C, Surarit R, Sila-Asna M, et al. Juice of Eclipta prostrata inhibits cell migration in vitro and exhibits anti-angiogenic activity in vivo. In Vivo. 2008;22:363-368
  94. 94. Ali F, Khan R, Khan AQ  , Lateef MA, Maqbool T, Sultana S. Assessment of augmented immune surveillance and tumor cell death by cytoplasmic stabilization of p53 as a chemopreventive strategy of 3 promising medicinal herbs in murine 2-stage skin carcinogenesis. Integrative Cancer Therapies. 2014;13:351-367
  95. 95. Dickers KJ, Bradberry SM, Rice P, Griffiths GD, Vale JA. Abrin poisoning. Toxicological Reviews. 2003;22:137-142
  96. 96. Inglett GE, May JF. Tropical plants with unusual taste properties. Economic Botany. 1968;22:326-331
  97. 97. Verma D, Tiwari SS, Srivastava S, Rawat A. Pharmacognostical evaluation and phytochemical standardization of Abrus precatorius L. seeds. Natural Product Sciences. 2011;17:51-57
  98. 98. Garaniya N, Bapodra A. Ethnobotanical and phytopharmacological potential of Abrus precatorius L.: A review. Asian Pacific Journal of Tropical Biomedicine. 2014;4(Suppl 1):S27-S34
  99. 99. Gadadhar S, Karande AA. Abrin immunotoxin: Targeted cytotoxicity and intracellular trafficking pathway. PLoS One. 2013;8(3):e58304
  100. 100. Panneerselvam K, Lin SC, Liu CL, Liaw YC, Lin JY, Lu TH. Crystallization of agglutinin from the seeds of Abrus precatorius. Acta Crystallographica. Section D, Biological Crystallography. 2000;56:898-899
  101. 101. Bhutia SK, Mallick SK, Stevens SM, Prokai L, Vishwanatha JK, Maiti TK. Induction of mitochondria-dependent apoptosis by Abrus agglutinin derived peptides in human cervical cancer cells. Toxicology In Vitro. 2008;22:344-351
  102. 102. Gul MZ, Ahmad F, Kondapi AK, Qureshi IA, Ghazi IA. Antioxidant and antiproliferative activities of Abrus precatorius leaf extracts—An in vitro study. BMC Complementary and Alternative Medicine. 2013;13:53. DOI: 10.1186/1472-6882-13-53
  103. 103. Lébri M, Tilaoui M, Bahi C, Achibat H, Akhramez S, Fofié YBN, et al. Phytochemical analysis and in vitro anticancer effect of aqueous extract of Abrus precatorius Linn. Der Pharma Chemica. 2015;7:112-117
  104. 104. Shafi Sofi M, Sateesh MK, Bashir M, Harish G, Lakshmeesha TR, Vedashree S, et al. Cytotoxic and pro-apoptotic effects of Abrus precatorius L. on human metastatic breast cancer cell line, MDA-MB-231. Cytotechnology. 2013;65:407-417
  105. 105. Reddy VV, Sirsi M. Effect of Abrus precatorius L. on experimental tumors. Cancer Research. 1969;29:1447-1451
  106. 106. Bhutia SK, Mallick SK, Maiti S, Maiti TK. Antitumor and proapoptotic effect of Abrus agglutinin derived peptide in Dalton's lymphoma tumor model. Chemico-Biological Interactions. 2008;174:11-18
  107. 107. Bhutia SK, Mallick SK, Maiti TK. In vitro immunostimulatory properties of Abrus lectins derived peptides in tumor bearing mice. Phytomedicine. 2009;16:776-782
  108. 108. Anbu J, Ravichandiran V, Sumithra M, Chowdary SB, Kumar S, Kannadhasan R, et al. Anticancer activity of petroleum ether extract of Abrus precatorius on Ehrlich ascitis carcinoma in mice. International Journal of Pharma and Bio Sciences. 2011;2:24-31
  109. 109. Shionoya H, Arai H, Koyanagi N, Ohtake S, Kobayashi H, Kodama T, et al. Induction of antitumor immunity by tumor cells treated with abrin. Cancer Research. 1982;42:2872-2876
  110. 110. Ramnath V, Kuttan G, Kuttan R. Immunopotentiating activity of abrin, a lectin from Abrus precatorius Linn. Indian Journal of Experimental Biology. 2002;40:910-913
  111. 111. Ghosh D, Bhutia SK, Mallick SK, Banerjee I, Maiti TK. Stimulation of murine B and T lymphocytes by native and heat-denatured Abrus agglutinin. Immunobiology. 2009;214:227-234
  112. 112. Van Andel T. The diverse uses of fish-poison plants in northwest Guyana. Economic Botany. 2000;54:500-512
  113. 113. Fukami J, Shishido T, Fukunaga K, Casida JE. Oxidative metabolism of rotenone in mammals, fish, and insects and its relation to selective toxicity. Journal of Agricultural and Food Chemistry. 1969;17:1217-1226
  114. 114. Qureshi R, Bhatti GR, Memon RA. Ethnomedicinal uses of herbs from northern part of NARA desert, Pakistan. Pakistan Journal of Botany. 2010;42:839-851
  115. 115. Vasconcelos JN, Lima JQ, de Lemos TLG, Oliveira MCF, Almeida MMB, Andrade-Neto M, et al. Estudo químico e biologico de Tephrosia toxicaria Pers. (Chemical and biological study of Tephrosia toxicaria Pers.). Quím Nova. 2009;32:382-386
  116. 116. Touqeer S, Saeed MA, Ajaib M. A review on the phytochemistry and pharmacology of genus Tephrosia. Phytopharmacology. 2013;4:598-637
  117. 117. Chen Y, Yan T, Gao C, Cao W, Huang R. Natural products from the genus Tephrosia. Molecules. 2014;19:1432-1458
  118. 118. Adinarayana K, Jayaveera KN, Madhu Katyayani B, Mallikarjuna Rao P. Growth inhibition and induction of apoptosis in estrogen receptor positive and negative human breast carcinoma cells by Tephrosia calophylla roots. Pharmaceutical Chemistry Journal. 2009;3:35-41
  119. 119. Gulecha V, Sivakuma T. Anticancer activity of Tephrosia purpurea and Ficus religiosa using MCF 7 cell lines. Asian Pacific Journal of Tropical Medicine. 2011;4:526-529
  120. 120. Nondo RS, Mbwambo ZH, Kidukuli AW, Innocent EM, Mihale MJ, Erasto P, et al. Larvicidal, antimicrobial and brine shrimp activities of extracts from Cissampelos mucronata and Tephrosia villosa from coast region, Tanzania. BMC Complementary and Alternative Medicine. 2011;11:33-37
  121. 121. Shanmugapriya R, Umamaheswari G, Thirunavukkarasu P, Renugadevi G, Ramanathan T. Cytotoxic effect of Tephrosia purpurea extracts on HeLa cervical cancerous cell line. Inventi Rapid: Molecular Pharmacology 2011;2. Article ID “Inventi:mp/49/11”
  122. 122. Subhadra S, Kanacharalapalli VR, Ravindran VK, Parre SK, Chintala S, Thatipally R. Comparative toxicity assessment of three Tephrosia species on Artemia salina and animal cell lines. Journal of Natural Pharmaceuticals. 2011;2:143-148
  123. 123. Khalighi-Sigaroodi F, Ahvazi M, Hadjiakhoondi A, Taghizadeh M, Yazdani D, Khalighi-Sigaroodi S, et al. Cytotoxicity and antioxidant activity of 23 plant species of Leguminosae family. Iranian Journal of Pharmaceutical Research. 2012;11:295-302
  124. 124. Padmapriya R, Gayathri L, Ronsard L, Akbarsha MA, Raveendran R. In vitro antiproliferative effect of Tephrosia purpurea on human hepatocellular carcinoma cells. Pharmacognosy Magazine. 2017;13(Suppl 1):S16-S21
  125. 125. Ganapaty S, Srilakshmi GVK, Pannakal ST, Rahman H, Laatsch H, Brun R. Cytotoxic benzil and coumestan derivatives from Tephrosia calophylla. Phytochemistry. 2009;70:95-99
  126. 126. Ganapaty S, Srilakshmi GVK, Thomas PS, Rajeswari NR, Ramakrishna S. Cytotoxicity and antiprotozoal activity of flavonoids from three Tephrosia species. Journal of Natural Remedies. 2009;9:202-208
  127. 127. Hassan LE, Ahamed MB, Majid AS, Baharetha HM, Muslim NS, Nassar ZD, et al. Correlation of antiangiogenic, antioxidant and cytotoxic activities of some Sudanese medicinal plants with phenolic and flavonoid contents. BMC Complementary and Alternative Medicine. 2014;14:406. DOI: 10.1186/1472-6882-14-406
  128. 128. Hassan LEA, Iqbal MA, Dahham SS, Tabana YM, Ahamed MBK, Majid AMSA. Colorectal, prostate and pancreas human cancers targeted bioassay-guided isolations and characterization of chemical constituents from Tephrosia apollinea. Anti-Cancer Agents in Medicinal Chemistry. 2017;17:590-598
  129. 129. Jang DS, Park EJ, Kang YH, Hawthorne ME, Vigo JS, Graham JG, et al. Potential cancer chemopreventive flavonoids from the stems of Tephrosia toxicaria. Journal of Natural Products. 2003;66:1166-1170
  130. 130. Saleem M, Ahmed S, Alam A, Sultana S. Tephrosia purpurea alleviates phorbol ester-induced tumor promotion response in murine skin. Pharmacological Research. 2001;43:135-144
  131. 131. Hussain T, Siddiqui HH, Fareed S, Vijayakumar M, Rao CV. Chemopreventive evaluation of Tephrosia purpurea against N-nitrosodiethylamine-induced hepatocarcinogenesis in Wistar rats. The Journal of Pharmacy and Pharmacology. 2012;64:1195-1205
  132. 132. Bright EO, Okusanya BA. Infestation of economic plants in Badeggi by Tapinanthus dodoneifolius (DC) Danser and Tapinanthus globiferus (A. Rich) Van Tiegh. Nigerian Journal of Weed Science. 1998;11:51-56
  133. 133. Roth I, Lindorf H. South American Medicinal Plants. Botany, Remedial Properties and General Use. Berlin: Springer Verlag; 2002
  134. 134. Yazbek PB, Tezoto J, Cassas F, Rodrigues E. Plants used during maternity, menstrual cycle and other women's health conditions among Brazilian cultures. Journal of Ethnopharmacology. 2016;179:310-331
  135. 135. Lindholm P. Cytotoxic compounds of plant origin—Biological and chemical diversity. [PhD thesis]. Uppsala: Faculty of Pharmacy; 2005
  136. 136. Mary KT, Girija K, Ramadasan K. Partial purification of tumour-reducing principle from Helicanthis elasticus (Fam. Loranthaceae). Cancer Letters. 1994;81:53-57
  137. 137. Fernandez T, Wagner ML, Varela BG, Ricco RA, Hajos SE, Gurni AA, et al. Study of an Argentine mistletoe, the hemiparasite Ligaria cuneifolia (R. et P.) Tiegh. (Loranthaceae). Journal of Ethnopharmacology. 1998;62:25-34
  138. 138. Winarno H. Antiproliferative activity of octadeca-8,10,12-triynoic acid against human cancer cell lines. Benia Biologi. 2009;9:343-348
  139. 139. Yoon TJ, Yoo YC, Kang TB, Shimazaki K, Song SK, Lee KH, et al. Lectins isolated from Korean mistletoe (Viscum album coloratum) induce apoptosis in tumor cells. Cancer Letters. 1999;136:33-40
  140. 140. Rios MY, Salina D, Villarreal ML. Cytotoxic activity of moronic acid and identification of the new triterpene 3,4-seco-olean-18-ene-3,28-dioic acid from Phoradendron reichenbachianum. Planta Medica. 2001;67:443-446
  141. 141. Sadik G, Islam R, Rahman MM, Khondkar P, Rashid MA, Sarker SD. Antimicrobial and cytotoxic constituents of Loranthus globosus. Fitoterapia. 2003;74:308-311
  142. 142. Kim YK, Kim YS, Choi SU, Ryu SY. Isolation of flavonol rhamnosides from Loranthus tanakae and cytotoxic effect of them on human tumor cell lines. Archives of Pharmacal Research. 2004;27:44-47
  143. 143. Zhao YL, Wang XY, Sun LX, Fan RH, Bi KS, Yu ZG. Cytotoxic constituents of Viscum coloratum. Zeitschrift für Naturforschung. Section C. 2012;67:129-134
  144. 144. Cerdá Zolezzi P, Fernández T, Aulicino P, Cavaliere V, Greczanik S, Caldas Lopes E, et al. Ligaria cuneifolia flavonoid fractions modulate cell growth of normal lymphocytes and tumor cells as well as multidrug resistant cells. Immunobiology. 2005;209:737-749
  145. 145. Murwani R. Indonesian tea mistletoe (Scurrula oortiana) stem extract increases tumour cell sensitivity to tumour necrosis factor alpha (TNFalpha). Phytotherapy Research. 2003;17:407-409
  146. 146. Elsyana V, Bintang M, Priosoeryanto BP. Cytotoxicity and antiproliferative activity assay of clove mistletoe (Dendrophthoe pentandra (L.) Miq.) leaves extracts. Advances in Pharmacological Sciences. 2016;2016:3242698. DOI: 10.1155/2016/3242698
  147. 147. Kumar N, Biswas S, Mathew AE, Varghese S, Mathew JE, Nandakumar K, et al. Pro-apoptotic and cytotoxic effects of enriched fraction of Elytranthe parasitica (L.) Danser against HepG2 hepatocellular carcinoma. BMC Complementary and Alternative Medicine. 2016;16:420
  148. 148. Ohashi K, Winarno H, Mukai M, Shibuya H. Preparation and cancer cell invasion inhibitory effects of C16-alkynic fatty acids. Chemical and Pharmaceutical Bulletin (Tokyo). 2003;51:463-466
  149. 149. Heitzman ME, Neto CC, Winiarz E, Vaisberg AJ, Hammond GB. Ethnobotany, phytochemistry and pharmacology of Uncaria (Rubiaceae). Phytochemistry. 2005;66:5-29
  150. 150. Sandoval M, Okuhama NN, Zhang XJ, Condezo LA, Lao J, Angeles FM, et al. Anti-inflammatory and antioxidant activities of cat's claw (Uncaria tomentosa and Uncaria guianensis) are independent of their alkaloid content. Phytomedicine. 2002;9:325-337
  151. 151. Lee KK, Zhou BN, Kingston DG, Vaisberg AJ, Hammond GB. Bioactive indole alkaloids from the bark of Uncaria guianensis. Planta Medica. 1999;65:759-760
  152. 152. Laus G. Advances in chemistry and bioactivity of the genus Uncaria. Phytotherapy Research. 2004;18:259-274
  153. 153. Miller MJ, Mehta K, Kunte S, Raut V, Gala J, Dhumale R, et al. Early relief of osteoarthritis symptoms with a natural mineral supplement and a herbomineral combination: A randomized controlled trial [ISRCTN38432711]. Journal of Inflammation (London). 2005;2:11
  154. 154. Mehta K, Gala J, Bhasale S, Naik S, Modak M, Thakur H, et al. Comparison of glucosamine sulfate and a polyherbal supplement for the relief of osteoarthritis of the knee: A randomized controlled trial [ISRCTN25438351]. BMC Complementary and Alternative Medicine. 2007;7:34
  155. 155. Mur E, Hartig F, Eibl G, Schirmer M. Randomized double blind trial of an extract from the pentacyclic alkaloid-chemotype of Uncaria tomentosa for the treatment of rheumatoid arthritis. The Journal of Rheumatology. 2002;29:678-681
  156. 156. Del Grossi Moura M, Lopes LC, Biavatti MW, Kennedy SA, de Oliveira E, Silva MC, et al. Oral herbal medicines marketed in Brazil for the treatment of osteoarthritis: A systematic review and meta-analysis. Phytotherapy Research. 2017;31:1676-1685
  157. 157. Bacher N, Tiefenthaler M, Sturm S, Stuppner H, Ausserlechner M, Kofler R. Oxindole alkaloids from Uncaria tomentosa induce apoptosis in proliferating, G0/G1-arrested and bcl-2-expressing acute lymphoblastic leukaemia cells. British Journal of Haematology. 2006;132:615-622
  158. 158. Garcia Prado E, Garcia Gimenez MD, De la Puerta Vazquez R, Espartero Sanchez JL, Saenz Rodriguez MT. Antiproliferative effects of mitraphylline, a pentacyclic oxindole alkaloid of Uncaria tomentosa on human glioma and neuroblastoma cell lines. Phytomedicine. 2007;14:280-284
  159. 159. García Giménez D, García Prado E, Sáenz Rodríguez T, Fernández Arche A, De la Puerta R. Cytotoxic effect of the pentacyclic oxindole alkaloid mitraphylline isolated from Uncaria tomentosa bark on human Ewing’s sarcoma and breast cancer cell lines. Planta Medica. 2010;76:133-136
  160. 160. Pilarski R, Filip B, Wietrzyk J, Kuras M, Gulewicz K. Anticancer activity of the Uncaria tomentosa DC. preparations with different oxindole alkaloid composition. Phytomedicine. 2010;17:1133-1139
  161. 161. Urdanibia I, Michelangeli F, Ruiz MC, Milano B, Taylor P. Anti-inflammatory and antitumoural effects of Uncaria guianensis bark. Journal of Ethnopharmacology. 2013;150:1154-1162
  162. 162. Budán F, Szabó I, Varjas T, Nowrasteh G, Dávid T, Gergely P, et al. Mixtures of Uncaria and Tabebuia extracts are potentially chemopreventive in CBA/Ca mice: A long-term experiment. Phytotherapy Research. 2011;25:493-500
  163. 163. De Paula LC, Fonseca F, Perazzo F, Cruz FM, Cubero D, Trufelli DC, et al. Uncaria tomentosa (cat's claw) improves quality of life in patients with advanced solid tumors. Journal of Alternative and Complementary Medicine. 2015;21:22-30
  164. 164. Araújo MCS, Farias ILG, Gutierres J, Dalmora SL, Flores N, Farias J, et al. Uncaria tomentosa—Adjuvant treatment for breast cancer: Clinical trial. Evidence-based Complementary and Alternative Medicine. 2012;2012:676984. DOI: 10.1155/2012/676984
  165. 165. Farias ILG, Araújo MCS, Farias JG, Rossato LV, Elsenbach LI, Dalmora SL, et al. Uncaria tomentosa for reducing side effects caused by chemotherapy in CRC patients: Clinical trial. Evidence-based Complementary and Alternative Medicine. 2012;2012:892182. DOI: 10.1155/2012/892182
  166. 166. Alves IABS, Miranda HM, Soares LAL, Randau KP. Simaroubaceae family: Botany, chemical composition and biological activities. Revista Brasileira de Farmacognosia. 2014;24:481-501
  167. 167. Almeida MMB, Arriaga AMC, Santos AKL, Lemos TLG, Braz-Filho R, Vieira IJC. Ocorrência e atividade biológica de quassinóides da última década (Occurrence and biological activity of quassinoids in the last decade). Quimica Nova. 2007;30:935-951
  168. 168. Vikas B, Akhil BS, Suja SR, Sujathan K. An exploration of phytochemicals from Simaroubaceae. Asian Pacific Journal of Cancer Prevention. 2017;18:1765-1767
  169. 169. Showalter HDH. Progress in the synthesis of canthine alkaloids and ring-truncated congeners. Journal of Natural Products. 2013;76:455-467
  170. 170. Diehl C, Ferrari A. Superiority of Quassia amara 4% cream over metronidazole 0.75% cream in the treatment of rosacea: A randomized, double-blinded trial. Journal of Clinical and Cosmetic Dermatology. 2017;1. DOI: 10.16966/2576-2826.117
  171. 171. Kupchan SM, Streelman DR. Quassimarin, a new antileukemic quassinoid from Quassia amara. The Journal of Organic Chemistry. 1976;41:3481-3482
  172. 172. Kitagawa I, Mahmud T, Yokota K, Nakagawa S, Mayumi T, Kobayashi M, et al. Indonesian medicinal plants. XVII. Characterization of quassinoids from the stems of Quassia indica. Chemical and Pharmaceutical Bulletin (Tokyo). 1996;44:2009-2014
  173. 173. Cachet N, Hoakwie F, Bertani S, Bourdy G, Deharo E, Stien D, et al. Antimalarial activity of simalikalactone E, a new quassinoid from Quassia amara L. (Simaroubaceae). Antimicrobial Agents and Chemotherapy. 2009;53:4393-4398
  174. 174. Houël E, Bertani S, Bourdy G, Deharo E, Jullian V, Valentin A, et al. Quassinoid constituents of Quassia amara L. leaf herbal tea. Impact on its antimalarial activity and cytotoxicity. Journal of Ethnopharmacology. 2009;126:114-118
  175. 175. Murakami C, Fukamiya N, Tamura S, Okano M, Bastow KF, Tokuda H, et al. Multidrug-resistant cancer cell susceptibility to cytotoxic quassinoids, and cancer chemopreventive effects of quassinoids and canthin alkaloids. Bioorganic & Medicinal Chemistry. 2004;12:4963-4968
  176. 176. Rivero-Cruz JF, Lezutekong R, Lobo-Echeverri T, Ito A, Mi Q, Chai HB, et al. Cytotoxic constituents of the twigs of Simarouba glauca collected from a plot in southern Florida. Phytotherapy Research. 2005;19:136-140
  177. 177. Jiang XM, Zhou Y. Canthin-6-one alkaloids from Picrasma quassioides and their cytotoxic activity. Journal of Asian Natural Products Research. 2008;10:1009-1012
  178. 178. Usami Y, Nakagawa-Goto K, Lang JY, Kim Y, Lai CY, Goto M, et al. Antitumor agents. 282. 2′-(R)-O-acetylglaucarubinone, a quassinoid from Odyendyea gabonensis as a potential anti-breast and anti-ovarian cancer agent. Journal of Natural Products. 2010;73:1553-1558
  179. 179. Shields M, Niazi U, Badal S, Yee T, Sutcliffe MJ, Delgoda R. Inhibition of CYP1A1 by quassinoids found in Picrasma excelsa. Planta Medica. 2009;75:137-141
  180. 180. Wiseman CL, Yap HY, Bedikian AY, Bodey GP, Blumenschein GR. Phase II trial of bruceantin in metastatic breast carcinoma. American Journal of Clinical Oncology. 1982;5:389-391
  181. 181. Arseneau JC, Wolter JM, Kuperminc M, Ruckdeschel JC. A Phase II study of bruceantin (NSC-165, 563) in advanced malignant melanoma. Investigational New Drugs. 1983;1:239-242
  182. 182. Shan GY, Zhang S, Li GW, Chen YS, Liu XA, Wang JK. Clinical evaluation of oral Fructus bruceae oil combined with radiotherapy for the treatment of esophageal cancer. Chinese Journal of Integrative Medicine. 2011;17:933-936
  183. 183. Grant KL, Lutz RB. Alternative therapies: Ginger. American Journal of Health-System Pharmacy. 2000;57:945-947
  184. 184. An K, Zhao D, Wang Z, Wu J, Xu Y, Xiao G. Comparison of different drying methods on Chinese ginger (Zingiber officinale Roscoe): Changes in volatiles, chemical profile, antioxidant properties, and microstructure. Food Chemistry. 2016;197(Part B:1292-1300
  185. 185. Rahmani AH, Shabrmi FM, Aly SM. Active ingredients of ginger as potential candidates in the prevention and treatment of diseases via modulation of biological activities. International Journal of Physiology, Pathophysiology and Pharmacology. 2014;6:125-136
  186. 186. Gupta R, Singh PK, Singh R, Singh RL. Pharmacological activities of Zingiber officinale (ginger) and its active ingredients: A review. International Journal of Innovation Science and Research. 2016;4:1-18
  187. 187. Lee E, Surh YJ. Induction of apoptosis in HL60 cells by pungent vanilloids, [6]-gingerol and [6]-paradol. Cancer Letters. 1998;134:163-168
  188. 188. Keum YS, Kim J, Lee KH, Park KK, Surh YJ, Lee JM, et al. Induction of apoptosis and caspase-3 activation by chemopreventive [6]-paradol and structurally related compounds in KB cells. Cancer Letters. 2002;177:41-47
  189. 189. Park YJ, Wen J, Bang S, Park SW, Song SY. [6]-Gingerol induces cell cycle arrest and cell death of mutant p53-expressing pancreatic cancer cells. Yonsei Medical Journal. 2006;47:688-697
  190. 190. Ishiguro K, Ando T, Maeda O, Ohmiya N, Niwa Y, Kadomatsu K, et al. Ginger ingredients reduce viability of gastric cancer cells via distinct mechanisms. Biochemical and Biophysical Research Communications. 2007;362:218-223
  191. 191. Rhode J, Fogoros S, Zick S, Wahl H, Griffith KA, Huang J, et al. Ginger inhibits cell growth and modulates angiogenic factors in ovarian cancer cells. BMC Complementary and Alternative Medicine. 2007;7:44. DOI: 10.1186/1472-6882-7-44
  192. 192. Kim JS, Lee SI, Park HW, Yang JH, Shin TY, Kim YC, et al. Cytotoxic components from the dried rhizomes of Zingiber officinale Roscoe. Archives of Pharmacal Research. 2008;31:415-418
  193. 193. Lee SH, Cekanova M, Baek SJ. Multiple mechanisms are involved in 6-gingerol-induced cell growth arrest and apoptosis in human colorectal cancer cells. Molecular Carcinogenesis. 2008;47:197-208
  194. 194. Brown AC, Shah C, Liu J, Pham JT, Zhang JG, Jadus MR. Ginger's (Zingiber officinale Roscoe) inhibition of rat colonic adenocarcinoma cells proliferation and angiogenesis in vitro. Phytotherapy Research. 2009;23:640-645
  195. 195. Sang S, Hong J, Wu H, Liu J, Yang CS, Pan MH, et al. Increased growth inhibitory effects on human cancer cells and anti-inflammatory potency of shogaols from Zingiber officinale relative to gingerols. Journal of Agricultural and Food Chemistry. 2009;57:10645-10650
  196. 196. Gan FF, Nagle AA, Ang X, Ho OH, Tan SH, Yang H, et al. Shogaols at proapoptotic concentrations induce G(2)/M arrest and aberrant mitotic cell death associated with tubulin aggregation. Apoptosis. 2011;16:856-867
  197. 197. Tan BS, Kang O, Mai CW, Tiong KH, Khoo AS, Pichika MR, et al. 6-Shogaol inhibits breast and colon cancer cell proliferation through activation of peroxisomal proliferator activated receptor gamma (PPARgamma). Cancer Letters. 2013;336:127-139
  198. 198. Surh YJ, Park KK, Chun KS, Lee LJ, Lee E, Lee SS. Antitumor promoting activities of selected pungent phenolic substances present in ginger. Journal of Environmental Pathology, Toxicology and Oncology. 1999;18:131-139
  199. 199. Kim M, Miyamoto S, Yasui Y, Oyama T, Murakami A, Tanaka T. Zerumbone, a tropical ginger sesquiterpene, inhibits colon and lung carcinogenesis in mice. International Journal of Cancer. 2009;124:264-271
  200. 200. Lee E, Park KK, Lee JM, Chun KS, Kang JY, Lee SS, et al. Suppression of mouse skin tumor promotion and induction of apoptosis in HL-60 cells by Alpina oxyphylla Miquel (Zingiberaceae). Carcinogenesis. 1998;19:1337-1381
  201. 201. Jeong CH, Bode AM, Pugliese A, Cho YY, Kim HG, Shim JH, et al. [6]-Gingerol suppresses colon cancer growth by targeting leukotriene A4 hydrolase. Cancer Research. 2009;69:5584-5591
  202. 202. Wu H, Hsieh MC, Lo CY, Liu CB, Sang S, Ho CT, et al. 6-Shogaol is more effective than 6-gingerol and curcumin in inhibiting 12-O-tetradecanoylphorbol 13-acetate induced tumor promotion in mice. Molecular Nutrition & Food Research. 2010;54:1296-1306
  203. 203. Vinothkumar R, Sudha M, Nalini N. Chemopreventive effect of zingerone against colon carcinogenesis induced by 1,2-dimethylhydrazine in rats. European Journal of Cancer Prevention. 2014;23:361-371
  204. 204. Ling H, Yang H, Tan SH, Chui WK, Chew EH. 6-Shogaol, an active constituent of ginger, inhibits breast cancer cell invasion by reducing matrix metalloproteinase-9 expression via blockade of nuclear factor-kappa B activation. British Journal of Pharmacology. 2010;161:1763-1777
  205. 205. Núñez-Sánchez MA, González-Sarrías A, Romo-Vaquero M, García-Villalba R, Selma MV, Tomás-Barberán FA, et al. Dietary phenolics against colorectal cancer—From promising preclinical results to poor translation into clinical trials: Pitfalls and future needs. Molecular Nutrition & Food Research. 2015;59:1274-1291
  206. 206. Marx WM, Teleni L, McCarthy AL, Vitetta L, McKavanagh D, Thomson D, et al. Ginger (Zingiber officinale) and chemotherapy-induced nausea and vomiting: A systematic literature review. Nutrition Reviews. 2013;71:245-254
  207. 207. Lin SR, Fu YS, Tsai MJ, Cheng H, Weng CF. Natural compounds from herbs that can potentially execute as autophagy inducers for cancer therapy. International Journal of Molecular Sciences. 2017;18:pii: E1412. DOI: 10.3390/ijms18071412
  208. 208. Ruela-de-Sousa RR, Fuhler GM, Blom N, Ferreira CV, Aoyama H, Peppelenbosch MP. Cytotoxicity of apigenin on leukemia cell lines: Implications for prevention and therapy. Cell Death & Disease. 2010;1:e19. DOI: 10.1038/cddis.2009.18
  209. 209. Tuorkey MJ. Molecular targets of luteolin in cancer. European Journal of Cancer Prevention. 2016;25:65-76
  210. 210. Yang CS, Wang H. Cancer preventive activities of tea catechins. Molecules. 2016;21:pii: E1679
  211. 211. Dybkowska E, Sadowska A, Świderski F, Rakowska R, Wysocka K. The occurrence of resveratrol in foodstuffs and its potential for supporting cancer prevention and treatment. A review. Roczniki Państwowego Zakładu Higieny. 2018;69:5-14
  212. 212. Bock PR, Hanisch J, Matthes H, Zänker KS. Targeting inflammation in cancer-related-fatigue: A rationale for mistletoe therapy as supportive care in colorectal cancer patients. Inflammation & Allergy Drug Targets. 2014;13:105-111
  213. 213. Baliga MS, Rao S, Rai MP, D’souza P. Radioprotective effects of the Ayurvedic medicinal plant Ocimum sanctum Linn. (holy basil): A memoir. Journal of Cancer Research and Therapeutics. 2016;12:20-27
  214. 214. Baharvand M, Jafari S, Mortazavi H. Herbs in oral mucositis. Journal of Clinical and Diagnostic Research. 2017;11:ZE05-ZE11
  215. 215. Yu J, Wang C, Kong Q, Wu X, Lu JJ, Chen X. Recent progress in doxorubicin-induced cardiotoxicity and protective potential of natural products. Phytomedicine. 2018;40:125-139
  216. 216. Chen H, Lin Z, Arnst KE, Miller DD, Li W. Tubulin inhibitor-based antibody-drug conjugates for cancer therapy. Molecules. 2017;22:1281. DOI: 10.3390/molecules22081281
  217. 217. Shankar Babu M, Mahanta S, Lakhter AJ, Hato T, Paul S, Naidu SR. Lapachol inhibits glycolysis in cancer cells by targeting pyruvate kinase M2. PLoS One. 2018;13:e0191419. DOI: 10.1371/journal.pone.0191419
  218. 218. Khalili M, Akbarzadeh A, Chiani M, Sepideh T. The effect of nanoliposomal and pegylated nanoliposomal forms of 6-gingerol on breast cancer cells. Research Journal of Recent Sciences. 2013;2:29-33
  219. 219. Cuendet M, Pezzuto JM. Antitumor activity of bruceantin: An old drug with new promise. Journal of Natural Products. 2004;67:269-272
  220. 220. Guo Z, Vangapandu S, Sindelar RW, Walker LA, Sindelar RD. Biologically active quassinoids and their chemistry: Potential leads for drug design. Frontiers in Medicinal Chemistry. 2009;4:285-308

Written By

Dennis R.A. Mans and Euridice R. Irving

Submitted: 06 August 2018 Reviewed: 27 October 2018 Published: 26 November 2018