Skip to main content
  • Research article
  • Open access
  • Published:

The number of intestinal bacteria is not critical for the enhancement of antitumor activity and reduction of intestinal toxicity of irinotecan by the Chinese herbal medicine PHY906 (KD018)

Abstract

Background

The four-herb Chinese medicine PHY906(KD018) has been shown to both enhance the in vivo antitumor activity of irinotecan (CPT-11) against colon cancer tumor allografts and alleviate intestinal toxicity caused by CPT-11.

Methods

Since intestinal bacteria can metabolize CPT-11 and PHY906, we investigated whether intestinal bacteria play a critical role in the in vivo activity of PHY906 in murine Colon-38 tumor-bearing mice. Intestinal bacteria were depleted using streptomycin/neomycin for 10 days before and during treatment with PHY906 and/or CPT-11. qPCR using 16S DNA group-specific primers was used to quantify the levels of the major intestinal bacteria.

Results

Both PHY906 and antibiotic treatment changed the profile of intestinal bacteria species: Lactobacillus/Enterococcus, Bacteroides, Clostridium leptum, and E. rectale/C. coccoides. Antibiotic treatment did not alter the ability of PHY906 to enhance the antitumor activity of CPT-11. Antibiotic treatment alone partially reduced animal body weight loss in CPT-11-treated mice. However, PHY906 treatment was able to protect against the body weight loss in the CPT-11/antibiotic treatment group. H&E and PCNA staining of intestine showed that antibiotic treatment partially reduced the intestinal damage caused by CPT-11 but not as effectively as PHY906 treatment. Antibiotic treatment plus PHY906 conferred the most effective protection of intestine histological structure against damage by CPT-11. Both PHY906 and antibiotic treatment inhibited CPT-11-associated inflammatory processes, including infiltration of the intestine by neutrophils, MCP1 and TNF-alpha mRNA expression in the intestine, and expression of pro-inflammatory cytokines G-CSF and MCP1 proteins in the plasma. However, whereas antibiotic treatment suppressed the mRNA expression of two important intestinal progenitor/stem cell markers, Olfm4 and Lgr5, PHY906 treatment resulted in enhanced expression of these two stem cell markers.

Conclusions

Alterations in the population of intestinal bacteria did not affect the abilities of PHY906 to enhance CPT-11 antitumor activity or reduce the intestinal toxicity associated with CPT-11 treatment. The major species of intestinal bacteria do not appear to play a role in PHY906’s enhancement of the therapeutic index of CPT-11 in tumor-bearing mice. Thus, patients with different intestinal bacterial profiles may still benefit from PHY906 treatment alongside CPT-11.

Peer Review reports

Background

PHY906 (KD018) is based on the formulation Huang Qin Tang (HQT), which consists of four main herbs: Glycyrrhiza uralensis Fisch (G), Paeonia lactiflora Pall (P), Scutellaria baicalensis Georgi (S), and Ziziphus jujuba Mill (Z). Huang Qin Tang has been used for over 1800 years to treat a variety of gastrointestinal symptoms including diarrhea, nausea and vomiting, and abdominal cramps. PHY906 is manufactured using high-quality herbs picked by experienced herbalists and manufactured following cGMP (current Good Manufacturing Practice). Consistent preparations of PHY906 have been made over a period of 10 years as documented by Phytomics QC using standardized chemical and biological fingerprints[1].

Irinotecan (CPT-11) in combination with 5-fluorouracil (5-FU) and leucovorin continues to be used as the first-line therapy for treatment of metastatic colorectal cancer (mCRC). CPT-11 is also approved as a second-line monotherapy for recurrent mCRC following treatment with an oxaliplatin-based regimen. The dose-limiting toxicities of CPT-11 include nausea and vomiting, abdominal pain/cramps, and diarrhea. CPT-11 administration has also been shown to cause gastrointestinal bleeding, a symptom often associated with colonic ulceration resulting from intestinal cell death and inflammation (3). Once CPT-11 is administrated, it is converted to the active metabolite SN-38 (7-Ethyl-10-hydroxy-camptothecin) by hepatic and intestinal carboxyesterases[24]. SN-38 damages DNA in intestinal cells as well as tumor cells, and can trigger acute life-threatening diarrhea in patients[5]. In the liver, SN-38 is metabolized by hepatic UDP–glucuronyltransferase into the inactive SN-38G metabolite, which is excreted into the jejunum via the bile duct[6, 7]. Intestinal bacterial β-glucuronidase can convert SN-38G back into the SN-38 metabolite, which can directly damage the intestine and promote intestinal inflammation. Several approaches have been used to reduce GI toxicity resulting from CPT-11-based chemotherapy, including modification of the schedule of administration[8, 9], intestinal alkalization[10, 11], treatment with anti-diarrheal therapies[1216], genetic testing[17], ABCB1 transporter inhibitor treatment[18, 19], treatment with enzyme (β-glucuronidase, UGT1A1, carboxylesterase, COX-2) inducers or inhibitors[2023], antibiotic treatment[2426], treatment with adsorbing agents[27, 28], and treatment with other agents[2932]. However, to date, none of these approaches has been found to be of significant clinical benefit.

Since Huang Qin Tang has been used for over 1800 years ago to treat diarrhea, PHY906 was chosen as a potential treatment strategy to reduce the GI side effects associated with CPT-11 treatment. In in vivo pre-clinical studies, PHY906 reduced the GI toxicities caused by CPT-11 while simultaneously enhancing CPT-11’s in vivo antitumor activity[33]. Each of the four main herbs comprising the PHY906 formula was required to maximize both its cytoprotective and antitumor effects. A phase 1/2 randomized double-blinded, placebo-controlled clinical trial was conducted to examine PHY906’s clinical potential as an adjuvant to CPT-11, 5-fluorouracil, and leucovorin (5FU/LV) for the treatment of chemotherapy-naive mCRC patients. PHY906 treatment resulted in a significant decrease in the incidence of nausea/vomiting and diarrhea, and of note, treatment with PHY906 was not associated with any toxicity on its own[34, 35]. The pharmacokinetics of CPT-11 and 5FU, respectively, was not affected by PHY906 treatment[34].

PHY906 is orally administrated, and the individual herbal components can be metabolized and transformed by gut flora. Individuals may carry different profiles of intestinal bacteria in their GI system depending on gender[36], diet[37], age[38], and co-morbid illnesses[38, 39]. Therefore, the impact of PHY906 may be different for individuals with different spectra of intestinal flora. To test this hypothesis, we investigated the potential interaction of PHY906 and CPT-11 on intestinal damage and antitumor activity in mice with or without depletion of intestinal bacteria. Our findings demonstrate that the ability of PHY906 to increase the therapeutic index of PHY906 is not affected by levels of major bacteria species in the intestine.

Methods

Murine Colon-38 cells were transplanted subcutaneously into four- to six-week-old female BDF1 mice (Charles River Laboratories, Wilmington, MA). After 10 to 14 days, mice with tumor sizes of 150–300 mm3 were selected. Intestinal bacteria were depleted using streptomycin/neomycin (200 mg/kg, p.o. bid) for 10 days before and during the treatment of PHY906 and/or CPT-11. PHY906 was given orally (p.o.) for four days (twice per day; b.i.d., 500 mg/kg) and CPT-11 (360 mg/kg) was administered intraperitioneally (i.p.) on day 0. On day 0, PHY906 was given 30 minutes prior to CPT-11 administration. Mice (BDF1 bearing Colon-38 tumors) were terminated by cervical dislocation on day 4. Total DNA of the middle jejunum and the colon were purified using DNeasy Blood & Tissue Kit (Qiagen, Germantown, MD) according to manufacturer's instructions. qPCR using 16S DNA group-specific primers for Eubacteria, Lactobacillus/Enterococcus group (Lact), Bacteroides group (Bact), Clostridium leptum group (Clept) and E. rectale/C. coccoides group (Erec) was used to quantify the major intestinal bacteria in the middle jejunum and the colon[40]. Middle jejunum tissues were removed, fixed in formalin, embedded in paraffin, and sectioned into 10 μm pieces. Immunohistochemistry was used to detect protein expression in the middle jejunum tissues. Apoptosis was quantified by cleaved caspase 3 staining[33]. Cell proliferation was determined by proliferating cell nuclear antigen (PCNA) staining[33]. Quantitative RT real-time PCR was used to quantify Lgr5, Olfm4, Bmi, TNFα, and MCP1 mRNA expression in the middle jejunum[33]. Cytokine expression in the plasma was measured using the BDTMCBA (Cytometric Bead Array) (BD biosciences, San Jose, CA) according to manufacturer's instructions[33]. Data were analyzed by one-way or two-way ANOVA (GraphPad Prism 5, San Diego, CA), Student’s T-test (Microsoft Office Excel), and correlation analysis (GraphPad Prism 4). Difference were considered to be statistically significant when P < 0.05. All animal experiments were carried out in accordance with an approved Yale University Institutional Animal Care and Use Committee (IACUC) protocol. Murine Colon 38 cell lines were provided by Dr. Giuseppe Pizzorno, Ph.D., Pharm.D (Translational Science, Nevada Cancer Institute, USA).

Results

Treatment with PHY906, antibiotics, and/or CPT-11 alters the profile of major intestinal bacteria species

Intestinal bacteria in Colon-38 tumor bearing BDF1 mice was depleted with streptomycin/neomycin (200 mg/kg, p.o. bid) for 10 days before (our preliminary result indicated 7-day pre-treatment was not sufficient to deplete intestinal bacteria, data not shown) treatment with PHY906 (500 mg, p.o. bid days 0–3) and/or CPT-11 (360 mg/kg, I.P. day 0). On day 4, qPCR was used to quantify the levels of major intestinal bacteria in the middle jejunum and in the colon, using 16S DNA group-specific primers for Eubacteria, Lactobacillus/Enterococcus group (Lact), Bacteroides group (Bact), Clostridium leptum group (Clept) and E. rectale/C. coccoides group (Erec). As seen in Figure 1, PHY906 treatment significantly decreased levels of Bact (P = 0.012) and Erec (P = 0.03) (Figure 1A, C, F) but caused a slight increase in Lact (Figure 1A, B). PHY906 treatment increased levels of Clept but not Lact, Bact, or Erec in the colon, and it had no effect on Clept levels in the middle jejunum (Figure 1A, I). CPT-11 treatment significantly reduced levels of Bact (P = 0.014) and Erec (P = 0.04) (Figure 1A, H, J) but not Lact or Clept in the middle jejunum. In the colon, CPT-11 increased the density of Lact (P = 0.03) (Figure 1A, G). Administration of PHY906 following CPT-11 treatment significantly increased levels of Clept (P = 0.04) and Erec (P = 0.03) (Figure 1A, I, J) but not Lact and Bact in the colon. In middle jejunum, PHY906 + CPT-11 treatment only increased the density of Erec (P = 0.03) (Figure 1A, F).

Figure 1
figure 1

Effect of antibiotics, PHY906, CPT-11 and PHY906/CPT-11(9/c) on intestinal bacteria densities in murine Colon-38 tumor-bearing BDF1 micex. (A) Quantitative PCR heat maps for Lactobacillus/Enterococcus (Lact), Bacteroides (Bact), Clostridium leptum (Clept) and E. rectale/C. coccoides (Erec). The abundance of bacteria is categorized from high (red) to low (green) as indicated by the bar on the right side of the table. The top of the bar (100%) indicates the highest density of each bacterial group at each row of the table. (B-J) Quantitative PCR results for Lact, Bact, Clept and Erec in the middle jejunum (MJ) and colon. Each spot represents a mean of two or three different quantitative real-time PCR experiments (triplicate samples of each; 5 animals in each treatment group). Student's t-test was used to determine whether differences between treatment groups were significant. Details of experimental procedures are given in Methods.

In the colon, antibiotic treatment depleted the density of Bact, Clept, and Erec by more than 99% but did not affect that of Lact (Figure 1A, G-J). In the middle jejunum, antibiotics reduced levels of Lact, Bact, and Clept by about 95% and decreased Erec by nearly 80% (Figure 1A-F). After antibiotic treatment, both PHY906 and CPT-11 increased the density of Erec ( P = 0.02 and 0.04, respectively) (Figure 1A, J) but not Lact, Bact, or Clept in the colon; antibiotic + drug treatment also had no effect on the levels of tested bacteria in the middle jejunum. It is interesting to note that treatment with CPT-11, antibiotic, and PHY906 caused a significant increase in the density of Lact (P = 0.004) (Figure 1A, B) in the middle jejunum but not the colon. The level of Lact in CPT11/PHY906/antibiotic-treated mice was nearly 10-fold higher (P = 0.004) than that observed in the control group in antibiotic treatment conditions (Figure 1A, B).

Taken together, these studies show that PHY906 and CPT-11 with or without antibiotic treatment have variable effects on bacterial profiles in different regions of the intestine. Moreover, the interaction between PHY906 and CPT-11 could change bacterial profiles in the different segments of intestine.

The effect of antibiotic treatment on the anti-tumor activities of CPT-11 and CPT-11/PHY906

As it has been shown previously, treatment with PHY906 alone did not affect Colon-38 tumor growth. However, PHY906 was able to significantly enhance the antitumor activity of CPT-11 against Colon-38 cells (P < 0.0083) (Figure 2A). Treatment with antibiotics alone did not alter Colon-38 tumor growth (Figure 2A). Furthermore, antibiotic treatment did not affect the antitumor activity of either PHY906 or CPT-11 (Figure 2A). Most importantly, antibiotic treatment had no effect on the ability of PHY906 to significantly enhance the antitumor activity of CPT-11 against Colon-38 tumor growth (P < 0.0001). Cleaved caspase-3 staining indicated that PHY906 increased the level of apoptosis in Colon-38 tumors induced by CPT-11 with (P = 0.014) or without (P = 0.002) antibiotic treatment (Figure 2B, C).

Figure 2
figure 2

Effect of antibiotic treatment on the anti-tumor activity and GI toxicity of PHY906, CPT-11 and PHY906/CPT-11(9/c) in murine Colon-38 tumor-bearing BDF1 mice. (A) Effect of PHY906 and/or CPT-11 on the growth of Colon 38 tumor cells in BDF1 mice with or without (w/o) antibiotic treatment. Each spot represents an individual tumor’s size relative to that at initial treatment. One way ANOVA was used to determine the significance of the differences between curves (B) Immunohistochemistry staining for cleaved caspase-3 in Colon-38 tumor sections under different treatment conditions as indicated on the graph. Photographs in the figure were taken at 200× magnification. (C) Quantitation of cleaved caspase-3 staining. Cleaved caspase-3 stained cell per each view of Colon-38 tumor sections following different treatment conditions as indicated on the graph. Each spot represents the mean number of heavily stained cells from 4 to 5 views of each tumor section. Student's t-test was used to determine whether differences between treatment groups were significant. Details of the experimental procedures are given in Methods.

PHY906 reduced GI damage caused by CPT-11 regardless of antibiotic treatment

PHY906 treatment did little to reverse the body weight loss caused by CPT-11 by day 2 but promoted a significant recovery in body weight by day 4 (Figure 3A), a result consistent with our previous findings[33]. Antibiotic treatment partially mitigated the body weight loss triggered by CPT-11 between day 2 and day 4 (P = 0.051) (Figure 3A). PHY906 further protected against CPT-11-triggered body weight loss regardless of antibiotic treatment (Figure 3A). There was no difference in animal body weight between the CPT-11/PHY906 group and the CPT-11/PHY906/antibiotic group on day 4 (Figure 3A); PHY906 alone could significantly protect against body weight loss following CPT-11 treatment on day 4 (Figure 3A). In conclusion, PHY906 was able to enhance the antitumor activity of CPT11 and protect against CPT11-triggered body weight loss in mice with different intestinal bacteria profiles.

Figure 3
figure 3

Effect of PHY906 on CPT-11-induced body weight loss and damage of the middle jejunum in murine Colon-38 tumor bearing BDF1 mice with or without (w/o) antibiotic treatment on day 4. (A) Effect of PHY906 in protecting against weight loss induced by CPT-11 with or without (w/o) antibiotic treatment. Error bars indicate standard deviations; N = 5. (B) Hematoxylin and eosin staining for visualizing the formalin fixed sections of the middle jejunum four days after treatments commenced. Photographs were taken at 200x magnification. (C) Effect of PHY906 on the expression of Proliferating Cell Nuclear Antigen (PCNA) following CPT-11 treatment with or without antibiotic treatment. (D-F) qPCR for the mRNA expression of the proposed intestinal stem cell markers Lgr5, Olfm4 and Bmi1 in the middle jejunum under different treatment conditions. β-actin was used as an internal control. Each spot represents the mean of two or three different experiments (triplicate samples of each; N = 5). Student's t-test was used to determine whether differences between treatment groups were significant. Details of experimental procedures are given in Methods.

Consistent with our previous report[33], mice treated with CPT-11/PHY906 displayed improved histology in the middle jejunum, especially in the crypt area, as compared to mice treated with CPT-11 alone (Figure 3B). PCNA staining also indicated that the majority of GI crypt cells in mice treated with CPT-11/PHY906 were actively proliferating (Figure 3C). Antibiotic treatment was able to protect some crypts from damage caused by CPT-11 but was not nearly as effective as PHY906 treatment alone (Figure 3B). PCNA staining showed only some crypt cells actively proliferating in mice treated with antibiotics plus CPT-11 (Figure 3C). PHY906 plus antibiotic treatment yielded the most pronounced improvement in the histology of the middle jejunum from damage caused by CPT-11 (Figure 3B). However, RT-qPCR results indicated that antibiotic treatment could affect two important intestinal progenitor/stem cell markers: Lgr5 and Olfm4 mRNA expression. Antibiotic treatment alone strongly inhibited Lgr5 mRNA expression (P = 0.001) to a level similar to that in CPT-11-treated mice (Figure 3D). However, antibiotic treatment did not affect PHY906’s induction of Lgr5 mRNA expression following CPT-11 treatment (P < 0.0001) (Figure 3D). Furthermore, although antibiotic treatment did not significantly inhibit Olfm4 or Bmi mRNA expression (Figure 3E, F), antibiotic treatment inhibited PHY906’s induction of Olfm4 mRNA expression following CPT-11 treatment (Figure 3E). These results suggest that antibiotic treatment or the profile of intestinal bacteria may in some manner affect the behavior of intestinal progenitor/stem cells.

Anti-inflammation activity of PHY906 against CPT-11 toxicity is not affected by antibiotic treatment

As stated previously[33], PHY906 treatment inhibited several inflammatory processes triggered by CPT-11 in the middle jejunum, including neutrophil infiltration of the intestine (Figure 4A) and stimulation of TNF-α and MCP1 mRNA expression (Figure 4B and C). In the plasma, PHY906 reduced expression of pro-inflammatory cytokines induced by CPT-11 such as MCP1, G-CSF, and IL6 (Figure 4D-F). Antibiotic treatment also exhibited substantial anti-inflammatory activity in CPT-11-treated animals. It should be noted that 10-day pre-treatment with antibiotic significantly lowered the basal level of intestinal inflammation as reflected by a reduction in TNF-α (P = 0.003) and MCP1 (P = 0.01) mRNA (Figure 4B and C). However, antibiotic treatment did not affect basal levels of pro-inflammatory cytokines in the plasma.

Figure 4
figure 4

Effect of PHY906 on CPT-11-induced inflammation in murine Colon-38 tumor-bearing BDF1 mice with or without (w/o) antibiotic treatment on day 4. (A) Neutrophil infiltration in the middle jejunum section after different treatments. Photographs were taken at 200x magnification. (B and C) qPCR for TNFα and MCP-1 in the middle jejunum after treatment. β-actin was used as an internal control. Each spot represents the mean from two or three different experiments (triplicate samples of each; N = 5). (D-F) Detection of TNF-α, G-CSF and IL6 protein in the plasma after different treatments. (Each spot represents the mean from triplicate samples of each plasma sample; N = 5). Student's t-test was used to determine whether differences between treatment groups were significant. Details of experimental procedures are given in Methods.

Discussion

The Chinese herbal medicine Huang-Qin Tang has a nearly 2000-year history of use for treatment of GI side effects such as diarrhea, nausea, vomiting, and abdominal cramps. However, the direct effect of Huang-Qin Tang on intestinal bacteria levels has never been formally investigated. Herein, we investigated the ability of PHY906, a standardized cGMP “Huang-Qin Tang” equivalent, to alter bacterial profiles differently in different segments of intestine. Our in vivo studies have shown that PHY906 does not act as a broad-spectrum antibiotic against the bacteria that were tested in our studies. For example, PHY906 treatment only inhibited Bact and Erec while antibiotic treatment decreased all kinds of tested bacteria in the middle jejunum. Thus, the underlying mechanism of Huang-Qin Tang for the treatment of diarrhea is completely different from that of anti-diarrheal antibiotics. However, one caveat is that PHY906 might still have antibiotic activity against certain bacteria that were not tested in the present study. Indeed, several studies have reported that individual herbs of PHY906 possess antibiotic activity. Different Chemicals of Paeonia lactiflora were found to have different activities against Bact[41]. It was found that human intestinal bacteria could transform flavonoids of Scutellaria baicalensis into aglycone-flavonoids, baicalein, oroxylin A, wogonin and norwogonin, each of which has different potencies against different types of intestinal bacteria[42]. Glycyrrhizol A and 6,8-diisoprenyl-5,7,4'-trihydroxyisoflavone (5) isolated form Glycyrrhiza uralensis were shown to exhibit potent antibacterial activity against Streptococcus mutants[43]. Crude ethanol extracts of Ziziphus jujuba fruits were also reported to exhibit antibacterial activity[44]. Future investigations, such as a comparison of the impacts of different herbal combinations of PHY906 on intestinal bacterial profiles, could help us to address which herb(s) or chemical(s) are responsible for which of PHY906’s antibiotic activities.

Previous studies have shown that treatment of PHY906 with E. coli β-glucuronidase alters the mixture’s bioactivities. PHY906 treated with β-glucuronidase was found to exhibit stronger Wnt3a potentiation activity and anti-TNFα activity but weaker anti-iNOS activity in vitro[33]. Thus, using antibiotics to deplete intestine bacteria, which have high β-glucuronidase activity, would be expected to have an antagonistic effect on PHY906’s biological activity. In addition, antibiotic treatment should also lead to reduced SN38 formation and damage of intestinal tissues. Streptomycin/neomycin treatment depleted over 90% of the major bacteria species in the gut. This finding is consistent with other reports in the literature[24, 45]. We observed that antibiotic treatment partially protected intestinal tissue from CPT-11 toxicity without enhancing CPT-11’s antitumor activity. In contrast, PHY906 was able to reduce the extent of GI damage while enhancing the antitumor activity of CPT-11. Thus, our studies suggest that PHY906 is able to maintain its biological activity in the presence of a wide range of intestinal bacteria and their respective β-glucuronidases. Perhaps human β-glucuronidase and UDP-glucuronosyltransferase (UGT), both of which are expressed in intestinal tissues, may impact PHY906 metabolism. In our phase I/ll clinical trial for treatment of mCRC patients, most plasma flavonoids from the orally administrated PHY906 were found to be glucuronidated, although some were sulfonated or methylated[46]. Our preliminary studies suggest that both human glucuronidase and UGT(s) play key roles in the metabolism of flavonoids in PHY906.

Treatment with either PHY906 or antibiotics may be able to reduce CPT-11-induced inflammation, including neutrophil infiltration of intestine, MCP1 and TNF-α mRNA expression in the intestine, and increased expression of pro-inflammatory cytokines G-CSF and MCP1 proteins in the plasma. However, it appears that PHY906 and antibiotics have different mechanism of action. It is known that intestinal bacteria are normally localized to the loose mucus and cannot penetrate the inner mucus layer[4749]. However, damage to the mucus layer by CPT-11 allows intestinal bacteria to come in contact with epithelial cells or enter the blood stream, thus triggering the inflammatory process. Antibiotic treatment significantly reduces the level of intestinal bacteria and reduces the likelihood of bacteria-induced inflammation following CPT-11 treatment. This could also explain why antibiotic treatment alone reduced the basal level of MCP1 and TNF-α mRNA expression. In contrast, PHY906 accelerates the repopulation of epithelial cells, which restore the mucus layer and thus prevent bacterial penetration. PHY906 also suppresses inflammation by targeting several key signaling pathways, including NF-κB, iNOS, and COX2. For these reasons, the combination of antibiotics with PHY906 appears to more effectively suppress CPT-11-triggered inflammation and preserve the intestinal histological structure than either component alone. However, while antibiotic treatment did not affect PHY906’s induction of Lgr5 expression, it did suppress expression of intestinal progenitor/stem cell markers and inhibit PHY906’s ability to increase Olfm4 and Bmi levels following CPT-11 treatment. Lgr5[50, 51], Olfm4[52], and Bmi[53] all play important roles in maintaining the growth of intestinal progenitor/stem cells in the crypts of different segments of the intestine; intestinal bacteria can thus modulate intestinal stem cell proliferation to maintain gut homeostasis via the JAK–STAT and JNK pathways as demonstrated in Drosophila[54]. Therefore, the combination of PHY906 and antibiotics may not be the optimal approach to protect intestinal tissues from damage caused by CPT-11 or other toxic agents.

Inflammatory bowel diseases (IBD), including ulcerative colitis and Crohn’s disease, are caused by an innate immune response to luminal microflora in individuals with a certain genetic disposition. A recent finding indicated that the density of Clept is significantly reduced in the fecal microbiota of patients with Crohn’s disease and ulcerative colitis[39]. Our results showed that PHY906 alone can increase the density of Clept in colonic tissue. PHY906 is also able to maintain Clept and Erec levels in the colon following CPT-11 treatment. Therefore, PHY906 may have potential benefit in treating IBD by restoring the density of Clept in the colon. In addition, PHY906 inhibits NF-κB, COX2, and iNOS, all of which play key roles in IBD[5557]. Thus, the potential use of PHY906 in treating IBD is worthy of further investigation.

Conclusions

Our studies have shown that the depletion of intestinal bacteria by antibiotics does not impact the ability of PHY906 to enhance CPT-11’s antitumor activity and to protect against CPT-11-induced intestinal toxicity. Patients with a range of intestinal bacterial profiles may thus benefit from the use of PHY906 as a modulator of CPT-11-based treatment.

Abbreviations

CPT-11:

Irinotecan

qPCR:

Quantitative real-time polymerase chain reaction

RT-qPCR:

Reverse transcription quantitative real-time polymerase chain reaction

Lact:

Eubacteria, Lactobacillus/Enterococcus group

Bact:

Bacteroides group

Clept:

Clostridium leptum group

Erec:

E. rectale/C. coccoides group

mCRC:

metastatic colorectal cancer

GI:

Gastrointestinal

cGMP:

current Good Manufacturing Practice

G:

Glycyrrhiza uralensis Fisch

P:

Paeonia lactiflora Pall

S:

Scutellaria baicalensis Georgi

Z:

Ziziphus jujuba Mill

MCP1:

Monocyte Chemoattractant Protein-1

G-CSF:

Granulocyte colony stimulating factor

iNOS:

Inducible nitric oxide synthase

IBD:

Inflammatory bowel disease.

References

  1. Tilton R, Paiva AA, Guan JQ, Marathe R, Jiang Z, van Eyndhoven W, Bjoraker J, Prusoff Z, Wang H, Liu SH, Cheng YC: A comprehensive platform for quality control of botanical drugs (PhytomicsQC): a case study of Huangqin Tang (HQT) and PHY906. Chin Med. 2010, 5: 30-10.1186/1749-8546-5-30.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Khanna R, Morton CL, Danks MK, Potter PM: Proficient metabolism of irinotecan by a human intestinal carboxylesterase. Cancer Res. 2000, 60 (17): 4725-4728.

    CAS  PubMed  Google Scholar 

  3. Morton CL, Wierdl M, Oliver L, Ma MK, Danks MK, Stewart CF, Eiseman JL, Potter PM: Activation of CPT-11 in mice: identification and analysis of a highly effective plasma esterase. Cancer Res. 2000, 60 (15): 4206-4210.

    CAS  PubMed  Google Scholar 

  4. Satoh T, Hosokawa M, Atsumi R, Suzuki W, Hakusui H, Nagai E: Metabolic activation of CPT-11, 7-ethyl-10-[4-(1-piperidino)-1- piperidino]carbonyloxycamptothecin, a novel antitumor agent, by carboxylesterase. Biol Pharm Bull. 1994, 17 (5): 662-664. 10.1248/bpb.17.662.

    Article  CAS  PubMed  Google Scholar 

  5. Ikuno N, Soda H, Watanabe M, Oka M: Irinotecan (CPT-11) and characteristic mucosal changes in the mouse ileum and cecum. J Natl Cancer Inst. 1995, 87 (24): 1876-1883. 10.1093/jnci/87.24.1876.

    Article  CAS  PubMed  Google Scholar 

  6. Rivory LP, Bowles MR, Robert J, Pond SM: Conversion of irinotecan (CPT-11) to its active metabolite, 7-ethyl-10-hydroxycamptothecin (SN-38), by human liver carboxylesterase. Biochem Pharmacol. 1996, 52 (7): 1103-1111. 10.1016/0006-2952(96)00457-1.

    Article  CAS  PubMed  Google Scholar 

  7. Humerickhouse R, Lohrbach K, Li L, Bosron WF, Dolan ME: Characterization of CPT-11 hydrolysis by human liver carboxylesterase isoforms hCE-1 and hCE-2. Cancer Res. 2000, 60 (5): 1189-1192.

    CAS  PubMed  Google Scholar 

  8. Takimoto CH, Morrison G, Harold N, Quinn M, Monahan BP, Band RA, Cottrell J, Guemei A, Llorens V, Hehman H, Ismail AS, Flemming D, Gosky DM, Hirota H, Berger SJ, Berger NA, Chen AP, Shapiro JD, Arbuck SG, Wright J, Hamilton JM, Allegra CJ, Grem JL: Phase I and pharmacologic study of irinotecan administered as a 96-hour infusion weekly to adult cancer patients. J Clin Oncol. 2000, 18 (3): 659-667.

    CAS  PubMed  Google Scholar 

  9. Herben VM, Schellens JH, Swart M, Gruia G, Vernillet L, Beijnen JH, ten Bokkel Huinink WW: Phase I and pharmacokinetic study of irinotecan administered as a low-dose, continuous intravenous infusion over 14 days in patients with malignant solid tumors. J Clin Oncol. 1999, 17 (6): 1897-1905.

    CAS  PubMed  Google Scholar 

  10. Valenti Moreno V, Brunet Vidal J, Manzano Alemany H, Salud Salvia A, Llobera Serentill M, Cabezas Montero I, Servitja Tormo S, Sopena Bert E, Guma Padro J: Prevention of irinotecan associated diarrhea by intestinal alkalization. A pilot study in gastrointestinal cancer patients. Clin Transl Oncol. 2006, 8 (3): 208-212. 10.1007/s12094-006-0012-1.

    Article  PubMed  Google Scholar 

  11. Tamura T, Yasutake K, Nishisaki H, Nakashima T, Horita K, Hirohata S, Ishii A, Hamano K, Aoyama N, Shirasaka D, Kamigaki T, Kasuga M: Prevention of irinotecan-induced diarrhea by oral sodium bicarbonate and influence on pharmacokinetics. Oncology. 2004, 67 (5–6): 327-337.

    CAS  PubMed  Google Scholar 

  12. Benson AB, Ajani JA, Catalano RB, Engelking C, Kornblau SM, Martenson JA, McCallum R, Mitchell EP, O'Dorisio TM, Vokes EE, Wadler S: Recommended guidelines for the treatment of cancer treatment-induced diarrhea. J Clin Oncol. 2004, 22 (14): 2918-2926. 10.1200/JCO.2004.04.132.

    Article  CAS  PubMed  Google Scholar 

  13. Rosenoff SH: Octreotide LAR resolves severe chemotherapy-induced diarrhoea (CID) and allows continuation of full-dose therapy. Eur J Cancer Care (Engl). 2004, 13 (4): 380-383. 10.1111/j.1365-2354.2004.00511.x.

    Article  CAS  Google Scholar 

  14. Ychou M, Douillard JY, Rougier P, Adenis A, Mousseau M, Dufour P, Wendling JL, Burki F, Mignard D, Marty M: Randomized comparison of prophylactic antidiarrheal treatment versus no prophylactic antidiarrheal treatment in patients receiving CPT-11 (irinotecan) for advanced 5-FU-resistant colorectal cancer: an open-label multicenter phase II study. Am J Clin Oncol. 2000, 23 (2): 143-148. 10.1097/00000421-200004000-00008.

    Article  CAS  PubMed  Google Scholar 

  15. Karthaus M, Ballo H, Abenhardt W, Steinmetz T, Geer T, Schimke J, Braumann D, Behrens R, Behringer D, Kindler M, Messmann H, Boeck HP, Greinwald R, Kleeberg U: Prospective, double-blind, placebo-controlled, multicenter, randomized phase III study with orally administered budesonide for prevention of irinotecan (CPT-11)-induced diarrhea in patients with advanced colorectal cancer. Oncology. 2005, 68 (4–6): 326-332.

    Article  CAS  PubMed  Google Scholar 

  16. Duffour J, Gourgou S, Seitz JF, Senesse P, Boutet O, Castera D, Kramar A, Ychou M: Efficacy of prophylactic anti-diarrhoeal treatment in patients receiving Campto for advanced colorectal cancer. Anticancer Res. 2002, 22 (6B): 3727-3731.

    CAS  PubMed  Google Scholar 

  17. Evaluation of Genomic Applications in P, Prevention Working G: Recommendations from the EGAPP Working Group: can UGT1A1 genotyping reduce morbidity and mortality in patients with metastatic colorectal cancer treated with irinotecan?. Genet Med. 2009, 11 (1): 15-20.

    Article  Google Scholar 

  18. Vasudev NS, Jagdev S, Anthoney DA, Seymour MT: Intravenous irinotecan plus oral ciclosporin. Clin Oncol. 2005, 17 (8): 646-649. 10.1016/j.clon.2005.06.009.

    Article  CAS  Google Scholar 

  19. Desai AA, Kindler HL, Taber D, Agamah E, Mani S, Wade-Oliver K, Ratain MJ, Vokes EE: Modulation of irinotecan with cyclosporine: a phase II trial in advanced colorectal cancer. Cancer Chemother Pharmacol. 2005, 56 (4): 421-426. 10.1007/s00280-005-1020-5.

    Article  CAS  PubMed  Google Scholar 

  20. Fakih MG, Rustum YM: Does celecoxib have a role in the treatment of patients with colorectal cancer?. Clin Colorectal Cancer. 2009, 8 (1): 11-14. 10.3816/CCC.2009.n.002.

    Article  CAS  PubMed  Google Scholar 

  21. Sakata Y, Suzuki H, Kamataki T: Preventive effect of TJ-14, a kampo (Chinese herb) medicine, on diarrhea induced by irinotecan hydrochloride (CPT-11). Gan to kagaku ryoho Cancer Chemother. 1994, 21 (8): 1241-1244.

    CAS  Google Scholar 

  22. Mori K, Kondo T, Kamiyama Y, Kano Y, Tominaga K: Preventive effect of Kampo medicine (Hangeshashin-to) against irinotecan-induced diarrhea in advanced non-small-cell lung cancer. Cancer Chemother Pharmacol. 2003, 51 (5): 403-406.

    CAS  PubMed  Google Scholar 

  23. Tobin PJ, Beale P, Noney L, Liddell S, Rivory LP, Clarke S: A pilot study on the safety of combining chrysin, a non-absorbable inducer of UGT1A1, and irinotecan (CPT-11) to treat metastatic colorectal cancer. Cancer Chemother Pharmacol. 2006, 57 (3): 309-316. 10.1007/s00280-005-0053-0.

    Article  CAS  PubMed  Google Scholar 

  24. Kehrer DF, Sparreboom A, Verweij J, de Bruijn P, Nierop CA, van de Schraaf J, Ruijgrok EJ, de Jonge MJ: Modulation of irinotecan-induced diarrhea by cotreatment with neomycin in cancer patients. Clin Cancer Res. 2001, 7 (5): 1136-1141.

    CAS  PubMed  Google Scholar 

  25. Alimonti A, Satta F, Pavese I, Burattini E, Zoffoli V, Vecchione A: Prevention of irinotecan plus 5-fluorouracil/leucovorin-induced diarrhoea by oral administration of neomycin plus bacitracin in first-line treatment of advanced colorectal cancer. Ann Oncol. 2003, 14 (5): 805-806. 10.1093/annonc/mdg192.

    Article  CAS  PubMed  Google Scholar 

  26. de Jong FA, Kehrer DF, Mathijssen RH, Creemers GJ, de Bruijn P, van Schaik RH, Planting AS, van der Gaast A, Eskens FA, Janssen JT, Ruit JB, Verweij J, Sparreboom A, de Jonge MJ: Prophylaxis of irinotecan-induced diarrhea with neomycin and potential role for UGT1A1*28 genotype screening: a double-blind, randomized, placebo-controlled study. Oncologist. 2006, 11 (8): 944-954. 10.1634/theoncologist.11-8-944.

    Article  CAS  PubMed  Google Scholar 

  27. Michael M, Brittain M, Nagai J, Feld R, Hedley D, Oza A, Siu L, Moore MJ: Phase II study of activated charcoal to prevent irinotecan-induced diarrhea. J Clin Oncol. 2004, 22 (21): 4410-4417. 10.1200/JCO.2004.11.125.

    Article  CAS  PubMed  Google Scholar 

  28. Sergio GC, Felix GM, Luis JV: Activated charcoal to prevent irinotecan-induced diarrhea in children. Pediatr Blood Cancer. 2008, 51 (1): 49-52. 10.1002/pbc.21491.

    Article  PubMed  Google Scholar 

  29. Govindarajan R: Irinotecan/thalidomide in metastatic colorectal cancer. Oncology (Williston Park). 2002, 16 (4 Suppl 3): 23-26.

    Google Scholar 

  30. Villalona-Calero M, Schaaf L, Phillips G, Otterson G, Panico K, Duan W, Kleiber B, Shah M, Young D, Wu WH, Kuhn J: Thalidomide and celecoxib as potential modulators of irinotecan's activity in cancer patients. Cancer Chemother Pharmacol. 2007, 59 (1): 23-33.

    Article  CAS  PubMed  Google Scholar 

  31. Wakelee H, Fisher GA: A phase I trial of irinotecan (CPT-11) with amifostine in patients with metastatic colorectal cancer. Investig New Drugs. 2005, 23 (3): 241-242. 10.1007/s10637-005-6732-1.

    Article  Google Scholar 

  32. Delioukina ML, Prager D, Parson M, Hecht JR, Rosen P, Rosen LS: Phase II trial of irinotecan in combination with amifostine in patients with advanced colorectal carcinoma. Cancer. 2002, 94 (8): 2174-2179. 10.1002/cncr.10432.

    Article  CAS  PubMed  Google Scholar 

  33. Lam W, Bussom S, Guan F, Jiang Z, Zhang W, Gullen EA, Liu SH, Cheng YC: The four-herb Chinese medicine PHY906 reduces chemotherapy-induced gastrointestinal toxicity. Sci Transl Med. 2010, 2 (45): 45-59.

    Article  Google Scholar 

  34. Farrell MP, Kummar S: Phase I/IIA randomized study of PHY906, a novel herbal agent, as a modulator of chemotherapy in patients with advanced colorectal cancer. Clin Colorectal Cancer. 2003, 2 (4): 253-256. 10.3816/CCC.2003.n.007.

    Article  CAS  PubMed  Google Scholar 

  35. Kummar S, Copur MS, Rose M, Wadler S, Stephenson J, O'Rourke M, Brenckman W, Tilton R, Liu SH, Jiang Z, Su T, Cheng YC, Chu E: A phase I study of the chinese herbal medicine PHY906 as a modulator of irinotecan-based chemotherapy in patients with advanced colorectal cancer. Clin Colorectal Cancer. 2011, 10 (2): 85-96. 10.1016/j.clcc.2011.03.003.

    Article  CAS  PubMed  Google Scholar 

  36. Yurkovetskiy L, Burrows M, Khan AA, Graham L, Volchkov P, Becker L, Antonopoulos D, Umesaki Y, Chervonsky AV: Gender bias in autoimmunity is influenced by microbiota. Immunity. 2013, 39 (2): 400-412. 10.1016/j.immuni.2013.08.013.

    Article  CAS  PubMed  Google Scholar 

  37. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA, Biddinger SB, Dutton RJ, Turnbaugh PJ: Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014, 505 (7484): 559-563.

    Article  CAS  PubMed  Google Scholar 

  38. Hopkins MJ, Sharp R, Macfarlane GT: Age and disease related changes in intestinal bacterial populations assessed by cell culture, 16S rRNA abundance, and community cellular fatty acid profiles. Gut. 2001, 48 (2): 198-205. 10.1136/gut.48.2.198.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kabeerdoss J, Sankaran V, Pugazhendhi S, Ramakrishna BS: Clostridium leptum group bacteria abundance and diversity in the fecal microbiota of patients with inflammatory bowel disease: a case–control study in India. BMC Gastroenterol. 2013, 13: 20-10.1186/1471-230X-13-20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Salzman NH, Hung K, Haribhai D, Chu H, Karlsson-Sjoberg J, Amir E, Teggatz P, Barman M, Hayward M, Eastwood D, Stoel M, Zhou Y, Sodergren E, Weinstock GM, Bevins CL, Williams CB, Bos NA: Enteric defensins are essential regulators of intestinal microbial ecology. Nat Immunol. 2010, 11 (1): 76-83. 10.1038/ni.1825.

    Article  CAS  PubMed  Google Scholar 

  41. Ngan LT, Moon JK, Kim JH, Shibamoto T, Ahn YJ: Growth-inhibiting effects of Paeonia lactiflora root steam distillate constituents and structurally related compounds on human intestinal bacteria. World J Microbiol Biotechnol. 2012, 28 (4): 1575-1583. 10.1007/s11274-011-0961-6.

    Article  CAS  PubMed  Google Scholar 

  42. Xing S, Wang M, Peng Y, Chen D, Li X: Simulated gastrointestinal tract metabolism and pharmacological activities of water extract of Scutellaria baicalensis roots. J Ethnopharmacol. 2014, 152 (1): 183-189. 10.1016/j.jep.2013.12.056.

    Article  CAS  PubMed  Google Scholar 

  43. He J, Chen L, Heber D, Shi W, Lu QY: Antibacterial compounds from Glycyrrhiza uralensis. J Nat Prod. 2006, 69 (1): 121-124. 10.1021/np058069d.

    Article  CAS  PubMed  Google Scholar 

  44. Daneshmand F, Zare-Zardini H, Tolueinia B, Hasani Z, Ghanbari T: Crude Extract from Ziziphus Jujuba Fruits, a Weapon against Pediatric Infectious Disease. Iran J Pediatr Hematol Oncol. 2013, 3 (1): 216-221.

    CAS  Google Scholar 

  45. Takasuna K, Hagiwara T, Hirohashi M, Kato M, Nomura M, Nagai E, Yokoi T, Kamataki T: Involvement of beta-glucuronidase in intestinal microflora in the intestinal toxicity of the antitumor camptothecin derivative irinotecan hydrochloride (CPT-11) in rats. Cancer Res. 1996, 56 (16): 3752-3757.

    CAS  PubMed  Google Scholar 

  46. Zhang W, Saif MW, Dutschman GE, Li X, Lam W, Bussom S, Jiang Z, Ye M, Chu E, Cheng YC: Identification of chemicals and their metabolites from PHY906, a Chinese medicine formulation, in the plasma of a patient treated with irinotecan and PHY906 using liquid chromatography/tandem mass spectrometry (LC/MS/MS). J Chromatogr A. 2010, 1217 (37): 5785-5793. 10.1016/j.chroma.2010.07.045.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Johansson ME, Phillipson M, Petersson J, Velcich A, Holm L, Hansson GC: The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria. Proc Natl Acad Sci U S A. 2008, 105 (39): 15064-15069. 10.1073/pnas.0803124105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Velcich A, Yang W, Heyer J, Fragale A, Nicholas C, Viani S, Kucherlapati R, Lipkin M, Yang K, Augenlicht L: Colorectal cancer in mice genetically deficient in the mucin Muc2. Science. 2002, 295 (5560): 1726-1729. 10.1126/science.1069094.

    Article  CAS  PubMed  Google Scholar 

  49. Van der Sluis M, De Koning BA, De Bruijn AC, Velcich A, Meijerink JP, Van Goudoever JB, Buller HA, Dekker J, Van Seuningen I, Renes IB, Einerhand AW: Muc2-deficient mice spontaneously develop colitis, indicating that MUC2 is critical for colonic protection. Gastroenterology. 2006, 131 (1): 117-129. 10.1053/j.gastro.2006.04.020.

    Article  CAS  PubMed  Google Scholar 

  50. Sato T, Vries RG, Snippert HJ, van de Wetering M, Barker N, Stange DE, van Es JH, Abo A, Kujala P, Peters PJ, Clevers H: Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009, 459 (7244): 262-265. 10.1038/nature07935.

    Article  CAS  PubMed  Google Scholar 

  51. Barker N, van Es JH, Kuipers J, Kujala P, van den Born M, Cozijnsen M, Haegebarth A, Korving J, Begthel H, Peters PJ, Clevers H: Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature. 2007, 449 (7165): 1003-1007. 10.1038/nature06196.

    Article  CAS  PubMed  Google Scholar 

  52. van der Flier LG, van Gijn ME, Hatzis P, Kujala P, Haegebarth A, Stange DE, Begthel H, van den Born M, Guryev V, Oving I, van Es JH, Barker N, Peters PJ, van de Wetering M, Clevers H: Transcription factor achaete scute-like 2 controls intestinal stem cell fate. Cell. 2009, 136 (5): 903-912. 10.1016/j.cell.2009.01.031.

    Article  CAS  PubMed  Google Scholar 

  53. Sangiorgi E, Capecchi MR: Bmi1 is expressed in vivo in intestinal stem cells. Nat Genet. 2008, 40 (7): 915-920. 10.1038/ng.165.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Buchon N, Broderick NA, Chakrabarti S, Lemaitre B: Invasive and indigenous microbiota impact intestinal stem cell activity through multiple pathways in Drosophila. Genes Dev. 2009, 23 (19): 2333-2344. 10.1101/gad.1827009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Atreya I, Atreya R, Neurath MF: NF-kappaB in inflammatory bowel disease. J Intern Med. 2008, 263 (6): 591-596. 10.1111/j.1365-2796.2008.01953.x.

    Article  CAS  PubMed  Google Scholar 

  56. Park YS: COX-2 inhibitors in inflammatory bowel disease: friends or foes?. Korean J Gastroenterol. 2007, 50 (6): 350-355.

    PubMed  Google Scholar 

  57. Kolios G, Valatas V, Ward SG: Nitric oxide in inflammatory bowel disease: a universal messenger in an unsolved puzzle. Immunology. 2004, 113 (4): 427-437. 10.1111/j.1365-2567.2004.01984.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Pre-publication history

Download references

Acknowledgment

This work was supported by the National Cancer Institute (NCI) (PO1CA154295-01A1), and from the National Center for Complementary and Alternative Medicine (NCCAM), NIH, USA. Yung-Chi Cheng is a fellow of the National Foundation for Cancer Research, USA.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Yung-Chi Cheng.

Additional information

Competing interests

Yung-Chi Cheng and Shwu-Huey Liu are the co-inventors of PHY906 for cancer treatment.

Authors' contributions

WL did qPCR, took immunohischemical staining photos, analyzed results and wrote the manuscript. ZJ did animal experiments. Fulan Guan did immunohistochemical staining. RH did qPCR. Shwu-Huey Liu provided PHY906 and discussed results. EC discussed results and wrote the manuscript. Y-CC designed experiments, analyzed results and wrote the manuscript. All authors read and approved the final manuscript.

Wing Lam, Zaoli Jiang, Fulan Guan contributed equally to this work.

Authors’ original submitted files for images

Rights and permissions

Open Access  This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.

The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

To view a copy of this licence, visit https://creativecommons.org/licenses/by/4.0/.

The Creative Commons Public Domain Dedication waiver (https://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lam, W., Jiang, Z., Guan, F. et al. The number of intestinal bacteria is not critical for the enhancement of antitumor activity and reduction of intestinal toxicity of irinotecan by the Chinese herbal medicine PHY906 (KD018). BMC Complement Altern Med 14, 490 (2014). https://doi.org/10.1186/1472-6882-14-490

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1472-6882-14-490

Keywords