Skip to main content
Medical Science Monitor: International Medical Journal of Experimental and Clinical Research logoLink to Medical Science Monitor: International Medical Journal of Experimental and Clinical Research
. 2025 Jun 25;31:e947268. doi: 10.12659/MSM.947268

Antitumor Alkaloids in Tibetan Corydalis: Chemical Diversity and Pharmacological Insights

Linguo Cao 1,A,B,E, Lijun Liu 2,C, Yanyan Wang 1,D, Jiaqing Liu 1,E, Zuowu Xi 3,E,F,G,
PMCID: PMC12207934  PMID: 40556304

Abstract

The plant genus Corydalis (Papaveraceae), widely used in traditional Tibetan medicine, comprises numerous species rich in bioactive alkaloids exhibiting significant antitumor activities. Recent pharmacological studies demonstrate that these alkaloids exert potent anti-cancer effects through diverse molecular mechanisms, including cell cycle arrest, induction of apoptosis, suppression of angiogenesis, and inhibition of metastasis. However, comprehensive reviews specifically addressing their antitumor efficacy are limited. This article systematically summarizes current advances in understanding the chemical diversity, pharmacological mechanisms, clinical potential, and quality control of antitumor alkaloids derived from Tibetan medicinal Corydalis plants, proposing future research directions to promote their integration into modern oncological therapeutics.

Keywords: Chemistry, Corydalis, Medicine, Papaveraceae

Introduction

The genus Corydalis (Papaveraceae) is one of the largest plant genera, comprising approximately 428 species worldwide [1,2]. Over 120 species are native to the Qinghai-Tibet Plateau, predominantly distributed in southwestern China [3,4]. Historically, Tibetan medicine has extensively utilized several Corydalis species, including C. decumbens, C. yanhusuo, and C. bungeana, as well as traditional Tibetan remedies such as “Silva,” “Rhegonba,” and “BaXiaGa” [5]. Tibetan medicinal texts describe the entire Corydalis plant as useful for alleviating fever, detoxification, pruritus relief, moistening the lungs, and treating cough [6].

Recent pharmacological research has identified diverse alkaloids as key bioactive constituents of Corydalis, possessing multiple therapeutic properties, including analgesic, anti-inflammatory, antibacterial, hepatoprotective, neuroprotective, and particularly anti-cancer activities [7,8]. Alkaloids extracted from Tibetan medicinal species of Corydalis have demonstrated significant efficacy against various malignant tumors through multifaceted mechanisms, including induction of apoptosis, cell cycle arrest, suppression of tumor angiogenesis, and inhibition of metastasis [9,10].

In clinical practice, Corydalis alkaloids have been widely used in traditional Chinese medicine (TCM) and Tibetan medicine for pain management, cardiovascular diseases, and neurological disorders. For instance, tetrahydropalmatine, a major alkaloid in C. yanhusuo, is the active ingredient in Rotundine Tablets, which are prescribed for chronic pain and insomnia due to their low addiction potential and minimal adverse effects [13]. Additionally, C. decumbens and C. edulis extracts have shown promise in alleviating symptoms of Alzheimer’s disease by inhibiting β-amyloid aggregation and oxidative stress [14]. Recent studies also highlight the antitumor potential of Corydalis alkaloids, such as dehydrocorydalin and sanguinarine, which induce apoptosis and inhibit angiogenesis in various cancer models [15,16].

Despite these advancements, over 90% of Corydalis species remain chemically uncharacterized, limiting their therapeutic potential and quality control [17]. Furthermore, while several reviews have summarized the phytochemistry and bioactivities of Corydalis alkaloids, including a recent systematic review on alkaloid constituents [11,12], focused narrative reviews specifically addressing the antitumor effects of alkaloids from Tibetan medicinal plants remain limited. Such a review is essential to enhance the understanding of these medicinal plants and facilitate their integration into modern oncological practices.

Therefore, this article comprehensively reviews the antitumor effects of alkaloid constituents of Tibetan medicinal plants of the genus Corydalis. It highlights the current state of knowledge regarding their chemical diversity, antitumor mechanisms, therapeutic potential, and identifies directions for future research. By bridging traditional knowledge and modern pharmacology, this review seeks to advance the development of Corydalis-derived therapies for cancer treatment.

The History and Future of Corydalis in Tibetan Medicine

The Tibetan region has historically been recognized as a significant repository of medicinal plants in China, with preliminary figures indicating the presence of over a thousand distinct varieties of wild medicinal plant resources in this area [7]. Among these, the genus Corydalis represents a particularly abundant botanical resource in Tibet. Notably, the utilization of Corydalis in Tibet dates back over 1300 years [8], serving both as a dietary supplement and a medicinal resource for the prevention and treatment of various ailments. In the early eighth century A.D., the Tibetan medical text Yue Wang Yao Zhen (Moon King’s Medical Examination) first recorded the use of Corydalis as a medicinal herb [6]. Subsequent Tibetan medical classics, such as the Crystal Beads Materia Medica, the Four Medical Tantras, Shel Gong Shel Phreng, and the Blue Beryl, have documented Corydalis multiple times, demonstrating the gradual recognition of its medicinal properties and clinical applications in Tibetan medicine.

Globally, the genus Corydalis comprises a total of 428 distinct species, with China hosting 298 of these species. Of these, 219 are endemic species grouped into 10 distinct sections within China. Notably, Tibet harbors 94 species of Corydalis, Yunnan has over 70 species [9], Sichuan accommodates 95 species [10], Qinghai hosts 26 species [6], and Gansu contains 55 species [3]. The medicinal use of Corydalis species in southwestern China has been extensively documented. In the Sichuan area, 43 species are used for medicinal purposes [11]. In Qinghai, Tibetan medical practitioners utilize over 20 species therapeutically, while in Tibet, traditional Tibetan medicine employs approximately 71 species. Notably, traditional Tibetan medical formulas such as Nine-flavored Tibetan Aster Pill, Ganlangling Pill, Niuhuang Thirteen-flavored Pill, and Zhituo Jiebai Pill include various species of Corydalis plants [3].

Among the medicinal resources of the genus Corydalis, there are 71 original plant species corresponding to 27 medicinal varieties in Table 1 [12]. However, only 8 of these are included in official standards such as the 2020 edition of the Chinese Pharmacopoeia and the Standards of Tibetan Medicine in Table 2. Common varieties include C. pygmaea, C. saxicola, C. edulis, and C. yanhusuo [13]. However, most standards only cover characteristics, microscopic identification, and physicochemical identification; the quality standards are not yet comprehensive. Corydalis medicinal materials are widely documented in Tibetan medical literature, such as the Moon King’s Medical Examination, the Four Medical Tantras, and the Jingzhu Materia Medica, which describe the original plants, their efficacy, and the parts used [4]. However, discrepancies such as “different names for the same substance” and “same name for different substances” indicate inconsistencies in the identification of original plants and medicinal names across various texts [14]. Existing quality standards for Corydalis medicinal materials have deficiencies, including the lack of content determination and extractive measurements, which affect their accurate evaluation in the market [15]. This is mainly due to insufficient basic research, complex variety origins, and differing regional usage habits.

Table 1.

The recorded documentation of Corydalis genus Tibetan medicinal materials.

No. Original plant and herbal collection Efficacy Distribution
1 C. trachycarpa, C. conspersa, C. mucronifera, C. gortschakovii, C. zadoiensis, C. melanoohlors, C. curviflora, C. hamata, C. inopinata, C. spathulata, C. chrysosphaera, C. scaberula Antipyretic, reduce swelling, relieve plague, promote blood circulation and remove blood stasis, detoxify, relieve pain, dispel wind and regulate Qi, etc. Qinghai-Tibet Plateau, Tibet, Qinghai, Sichuan
2 C. scaberula, C. linarioides, C. densispica, C. rheinbabeniana, C. eugeniae, C. trifoliata Treating flu, pestilence, clearing heat and cooling blood, promoting blood circulation, and relieving pain Sichuan, Qinghai, Tibet
3 C. nigroapiculata, C. scaberula, C. moorcroftiana, C. kokiana, C. orthopoda, C. trachycarpa var. leucostachya Reducing fever, stopping bleeding, promoting blood circulation, and brightening the eyes Tibet, Qinghai
4 C. melanochlora, C. builifera, C. oxypetala subsp. balfouriana, C. conspersa, C. curviflora, C. cashmeriana, C. pubicaulis, C. straminea Treating epidemic diseases, clearing hidden heat, blood heat, etc. Sichuan, Qinghai, Tibet, Kashmir
5 C. melanochlora, C. builifera Clearing pulse heat, relieving pain, promoting blood circulation and removing blood stasis, dispelling wind and brightening the eyes Tibet, Qinghai
6 C. dasyptera, C. livida Clearing sore heat, treating poisonous sores, promoting new muscle growth, and removing scars and warts Tibet, Sichuan, Qinghai
7 C. adunca, C. stricta Clearing heat and detoxifying, soothing the liver and gallbladder, relieving pain and diarrhea Sichuan, Qinghai
8 C. bungeana, C. edulis Treating heat diseases, clearing heat and detoxifying, soothing the liver and gallbladder Qinghai, Tibet
9 C. tibetoalpina, C. lhorongensi, C. edulis Relieving fever, promoting blood circulation and removing blood stasis, clearing hidden heat Tibet, Qinghai
10 C. hamata Relieving fever, promoting blood circulation and removing blood stasis, dispelling wind and brightening the eyes Tibet

C. – Corydalis.

Table 2.

Overview of medicinal Corydalis species: Distribution, medicinal parts, included standards, and identification methods.

No. Drug Distribution area Medicinal part Included standards Identification items
1 C. hendersonii Qinghai, Tibet Whole plant Standards of Tibetan Medicine (1979), Ministry-issued Standards – Tibetan Medicine Macroscopic and Microscopic Identification
2 C. scaberula Sichuan, Tibet Whole plant Sichuan Medicinal Material Standards Macroscopic and Powder Identification
3 C. impatiens Qinghai, Sichuan, Gansu Whole plant Standards for Chinese Medicinal Materials of Sichuan Province (2010), Standards for Tibetan Medicinal Materials of Qinghai Province (2019) Macroscopic, Powder, and Physicochemical Identification
4 C. dasyptera Qinghai, Sichuan Whole plant Standards for Tibetan Medicinal Materials of Sichuan Province (2020) Macroscopic, Microscopic, and Thin-layer Chromatography Identification
5 C. melanochlora Tibet, Qinghai Whole plant Standards of Tibetan Medicine, etc. Macroscopic and Microscopic Identification
6 C. curviflora Sichuan, Tibet Whole plant Standards for Tibetan Medicinal Materials of Sichuan Province (2020); Standards of Tibetan Medicine, etc. Macroscopic and Microscopic Identification
7 C. bungeana Widely distributed nationwide Whole plant Chinese Pharmacopoeia 2020 Macroscopic, Microscopic, and Thin-layer Chromatography Identification
8 C. yanhusuo Sichuan, Yunnan Tuber Chinese Pharmacopoeia 2020 Macroscopic, Microscopic, and Thin-layer Chromatography Identification

C. – Corydalis.

In future research, it is necessary to improve quality standards by formulating more comprehensive criteria – including extractive measurements and content determination of key components – to enhance the controllability of medicinal quality. Simultaneously, the research and standardization of original plants should be strengthened by ensuring consistency between medicinal varieties and original plants through origin identification and chemical composition studies. Furthermore, conducting multi-component synergistic research to deeply explore active components beyond alkaloids will fully reveal their pharmacological effects and mechanisms of action. Through these efforts, it is expected to promote the rational utilization of Corydalis Tibetan medicinal resources, ensure their safe and standardized clinical use, and facilitate their further development in the field of modern medicine.

Phytochemical Research

The genus Corydalis is rich in various chemical constituents, including alkaloids, flavonoids, triterpenes, and volatile oils. Figure 1A illustrates the types of these constituents and their quantities within the genus Corydalis. To date, approximately 500 alkaloids have been identified and extracted from various species of the genus Corydalis. Specifically, 112 alkaloids have been isolated from 71 species of Tibetan medicinal Corydalis, as detailed in Table 3. These alkaloids encompass more than 10 different structural types, including protopine alkaloids (I), protoberberine alkaloids (II), phthalideisoquinoline alkaloids (III), benzylisoquinoline alkaloids (IV), spirobenzylisoquinoline alkaloids (V), aporphine alkaloids (VI), phenanthrene alkaloids (VII), cularine alkaloids (VIII), quinoline alkaloids (IX), and others (X) (Figure 1B). Among these, protoberberine alkaloids are the main chemical constituents in Tibetan medicinal Corydalis. In contrast, cularine alkaloids are rare and have a limited distribution, which is a distinctive feature of the genus Corydalis [3].

Figure 1.

Figure 1

Number and main structure of plant compounds in Corydalis. (A) The types and quantities of. various, compounds in Corydalis; (B) Corydalis is the main structural type of alkaloids in Tibetan medicine. Figure were created using Adobe Illustrator CC (Version 26.0, Adobe, Inc., USA).

Table 3.

Alkaloids isolated from Corydalis species employed as Tibetan medicines.

No Compound CAS Plant origin Structure type Ref.
1 13-oxoprotopine 15211-02-6 C. crispa I [16]
2 Cryptopine 482-74-6 C. hendersonii, C. ophiocarpa I [17]
3 Izmirine 89117-94-2 C. hendersonii I [18]
4 Protopine 130-86-9 C. mpatiens, C. hendersonii, C. ongipes, C. thyrsiflora, C. ophiocarpa, C. adunca, C. mucronifera, C. crispa, C. racemosa I [1921]
5 Pseudoprotopine 24240-05-9 C. racemosa I [22,23]
6 α-Allocryptopine 485-91-6 C. hendersonii, C. ophiocarpa I [24]
7 β-Allocryptopine 24240-04-8 C. hendersonii, C. ongipes I [25]
8 (−)-Corypalmine 6018-40-2 C. ophiocarpa II [26]
9 (−) -Corycarpinc C. ophiocarpa II [26]
10 (+)-Cavidine 32728-75-9 C. impatiens, C. thyrsiflora II [27]
11 (+)-Sinacthe C. meifolia II [28]
12 8-Oxycoptisine 19716-61-1 C. trachycarpa II [29]
13 9, 10-dihydroxy-2, 3-dimethoxytetrahydroprotoberberine C. taliensis II [30]
14 Apocavidine 32728-76-0 C. impatiens II [31,32]
15 Berberine 131-10-2 C. ophiocarpa II [26]
16 Canadine 522-97-4 C. hendersonii, C. ophiocarpa II [26]
17 Cheilanthifoline 483-44-3 C. impatiens, C. ophiocarpa, C. hendersonii, C. mucronifera, C. meifolia II [26,32]
18 Coptisine 3486-66-6 C. ophiocarpa, C. adunca, C. dasypterm II [26]
19 Coreximine 6719-49-9 C. crispa II [2]
20 Dehydrocavidine 83218-34-2 C. meifolia II [32,33]
21 Dehydrocheilanthifoline 38691-95-1 C. ophiocarpa II [34]
22 Dehydroisocorypalmine 3621-36-1 C. ophiocarpa II [26]
23 Isoapocavidine 616213-11-7 C. impatiens, C. meifolia II [20]
24 Palmatine 3486-67-7 C. adunca II [35]
25 Racemosine A C. racemosa II [23]
26 Scoulerine 6451-73-6 C. hendersonii II [36]
27 Sinactine 522-96-3 C. hendersonii, C. thyrsiflora II [36]
28 Tetrahydroberberrubine 17388-17-9 C. mucronifera II [21]
29 Tetrahydrocolumbamine 483-34-1 C. ophiocarpa, C. adunca, C. saxicola II [26,37]
30 Tetrahydrocoptisine 4312-32-7 C. impatiens, C. thyrsiflora, C. dasypterm, C. hendersonii, C. ophiocarpa II [23]
31 Tetrahydrocorysamine 32043-26-8 C. impatiens, C. dasypterm II [38]
32 Tetrahydroepiberberine 38853-67-7 C. impatiens II [20]
33 Tetrahydropalmatine 2934-97-6 C. adunca II [39]
34 Tetrahydrothalifendine 4668-22-8 C. impatiens, C. taliensis, C. hendersonii II [40]
35 (+)-Bicuculline 485-49-4 C. crispa III [40]
36 (S)-Stylopine 84-39-9 C. crispa III [41]
37 (±)-hypecorinine 117020-73-2 C. mucronifera III [21]
38 9-methyl-decumbenine C 1166335-84-7 C. hendersonii III [42]
39 Adlumine 38184-69-9 C. ophiocarpa, C. lineariodes III [21]
40 Bicuculline 485-49-4 C. impatiens, C. thyrsiflora, C. mucronifera III [20]
41 Capnoidine 485-50-7 C. impatiens III [43]
42 Corlumidine 25344-54-1 C. lineariodes III [44]
43 Corlumine 485-51-8 C. thyrsiflora III [32]
44 Hydrastine 118-08-1 C. stricta III [45]
45 6-acetonylacetylcorynoline C. taliensis IV [35,46]
46 6-Acetylambinine 96935-27-2 C. curviflora IV [47]
47 8-acetonydihydrosan- guinarine (6-acetonyl-5, 6-dihydrosanguinarine) C. hendersonii, C. thyrsiflora, C. adunca IV [35]
48 Acetylcorynoline 18797-80-3 C. taliensis, C. conspersa, C. melanochlora IV [48]
49 Ambiguanine A 1433460-91-3 C. curviflora IV [47]
50 Ambiguanine B 1433460-98-0 C. curviflora IV [47]
51 Ambiguanine C 1621607-90-6 C. curviflora IV [47]
52 Corynoline 18797-79-0 C. taliensis, C. longipes var. pubescens, C. adunca, C. conspersa, C. melanochlora, C. lineariodes IV [49]
53 Curviflorain A 2488784-48-9 C. curviflora IV [47]
54 Curviflorain B 2488784-49-0 C. curviflora IV [47]
55 Dihydrosanguinarine 3606-45-9 C. thyrsiflora, C. adunca, C. meifolia,C. mucronifera IV [21]
56 Isocorynoline 475-67-2 C. taliensis IV [26]
57 Sanguinarine 2447-54-3 C. ophiocarpa, C. stricta IV [26]
58 (S)-bulocapnine C. taliensis V [30]
59 Cassythicine 5890-28-8 C. impatiens V [40]
60 Corytuberine 517-56-6 C. dasypterm V [23]
61 Magnoflorine 2141/9/5 C. aliensis, C. racemosa V [30]
62 N-methylcalycinine 86537-66-8 C. taliensis V [40]
63 Corysolidine 56816-35-4 C. curviflora VI [47]
64 Norochotensimine 1292770-04-7 C. impatiens VI [40]
65 Ochotensimine 4829-36-1 C. impatiens, C. edulis VI [50]
66 Ochrobirine 24181-64-4 C. thyrsiflora, C. impatiens VI [2]
67 Sibiricine 64397-10-0 C. taliensis, C. hyrsiflora, C. mucronifera VI [34,51]
68 Yenhusomidine 56435-44-0 C. meifolia VI [32]
69 Yenhusomine C. meifolia VI [32]
70 (−)-oblongine 60008-01-7 C. taliensis VII [32]
71 (+)-Reticuline 485-19-8 C. taliensis VII [32]
72 1, 2, 3, 4-tet-rahydro-6-methoxy-1-[4-hydroxyphenyl]methyl-7-isoquinolinol C. adunca VII [52]
73 1, 2, 3, 4-tet-rahydro-7-methoxy-1-[4-hydroxyphenyl]methyl-8-isoquinolinol C. adunca VII [53]
74 Henderine 102686-11-3 C. longipes var. pubescens VII [53]
75 Taliensineside 4668-22-8 C. taliensis VII [14]
76 (−)-Pallidine 25650-75-3 C. racemosa VIII [23]
77 (−)-Sinoacutine 4090-18-0 C. racemosa VIII [23]
78 Corysamine 30243-28-8 C. dasypterm VIII [54]
79 Sibiricine 64397-10-0 C. crispa VIII [14]
80 13-oxocryptopine 15215-66-4 C. crispa IX [50]
81 Noroxyhydrastinine 21796-14-5 C. racemosa IX [23]
82 Oxohydrastinine 552-29-4 C. mucronifera IX [51]
83 Indole-3-carbadehyde 487-89-8 C. racemosa X [23]
84 Pyrrolezanthine 500574-37-8 C. racemosa XI [23]
85 (+)-Humosine A 11014-02-1 C. mucronifera XII [51]
86 (−)-magnocurarine 6801-40-7 C. racemosa XII [16]
87 (−)-7′-O-methylegenine C. mucronifera XII [51]
88 (±)corynoline 68035-45-0 C. trachycarpa XII [55]
89 1,1-dimethyl-6-methoxy-7-hydroxyl-1,2,3,4-tetrahydroisoquinoline C. curviflora XII [47]
90 6, 7-methylenedioxy-1(2H)-isoquinolinonenoroxyhydrastinine C. hendersonii XII [36]
91 8-methoxydihydrosanguinarine 1194396-25-2 C. mucronifera XII [21]
92 Amprotropine 134-53-2 C. delavayi XII [23]
93 Corynoxine 6877-32-3 C. conspersa XII [14]
94 Corypalline 450-14-6 C. ophiocarpa XII [56]
95 Coryximine 127460-61-1 C. curviflora XII [47]
96 Dehydrocorypalline 72142-82-6 C. ophiocarpa XII [57]
97 Fumaramine 30341-99-2 C. thyrsiflora XII [23]
98 Hendersine B 2035503-96-7 C. curviflora XII [47]
99 Isochotensine C. curviflora XII [47]
100 Jatrorrhizine 3621-38-3 C. racemosa XII [23]
101 Maclekarpine E 1228559-79-2 C. racemosa XII [23]
102 Mucroniferanine A 2218471-48-6 C. mucronifera XII [21]
103 Mucroniferanine B 2218471-49-7 C. mucronifera XII [21]
104 Mucroniferanine C 2218471-50-0 C. mucronifera XII [21]
105 Mucroniferanine D 2218471-51-1 C. mucronifera XII [21]
106 Mucroniferanine E 2218471-52-2 C. mucronifera XII [21]
107 Mucroniferanine F 2218471-53-3 C. mucronifera XII [21]
108 Mucroniferanine G 2218471-54-4 C. mucronifera XII [21]
109 Ochotensine 4959-88-0 C. thyrsiflora XII [52]
110 Racemosine B C. racemosa XII [23]
111 Rhoeagenine 5574-77-6 C. crispa XII [2]
112 Thalifendine 18207-71-1 C. racemosa XII [23]

I. Proto-opioid alkaloids; II. Berberine alkaloids; III. Berberine alkaloids; IV. Benzylisoquinoline alkaloids; V. Spirobenzylisoquinoline alkaloids; VI. Benzylisoquinoline alkaloids; VII. Apophenoid alkaloids; VIII. Phenanthrene alkaloids; IX. Curalin alkaloids; X. Indole alkaloids; XI. Pyrrole alkaloids; XII. Other alkaloids. C. – Corydalis.

Advances and Perspectives in Multidimensional Quality Marker (Q-Marker) Systems for Tibetan Medicinal Plants of the Genus Corydalis

Plants of the genus Corydalis (Papaveraceae), integral to Tibetan medicinal systems, present a critical bridge between traditional therapeutic wisdom and modern scientific validation. Recent advances in the theory of Quality Markers (Q-markers) have shifted research paradigms from single-component quantification to multidimensional, systemic quality evaluation frameworks. This review synthesizes progress and challenges in Q-marker identification for Tibetan Corydalis species across 6 dimensions: pharmacological activity networks, phytochemical profiling, analytical innovation, processing-induced dynamics, geo-authenticity fingerprints, and formula compatibility mechanisms.

Pharmacologically Guided Q-Marker Discovery: From Compound Identification to Mechanistic Elucidation

The analgesic and cardiovascular regulatory properties of Corydalis species, as documented in Tibetan medical classics, have been mechanistically validated. Tetrahydropalmatine (THP), a core bioactive alkaloid, exhibits superior analgesic potency (ED50: 8.7–12.3 mg/kg) in neuropathic pain models compared to morphine (ED50: 6.5 mg/kg) while lacking addictive liability, molecularly decoding the “non-addictive analgesia” described in the Four Medical Tantras [22,45]. Dehydrocorydaline, another key alkaloid, suppresses NLRP3 inflammasome activation, achieving 68% inhibition of platelet aggregation and 42% reduction in atherosclerotic plaque area [66,67], thereby demystifying the “blood-activating and stasis-resolving” effects. Notably, the multitarget nature of Corydalis alkaloids is increasingly recognized: protopine modulates both vascular NO/cGMP signaling (EC50: 4.2 μM) and central GABA_A receptors (Ki: 0.8 μM), aligning with the Tibetan holistic principle of “simultaneous somatic and psychic regulation” [69,72]. These breakthroughs not only validate alkaloids as Q-markers but also establish a robust “constituent-target-phenotype” evidence chain.

Chemotaxonomic Profiling: From Species Differentiation to Geo-Authenticity Assessment

The global chemodiversity of 428 Corydalis species poses challenges for quality standardization. UPLC-QTOF-MS metabolomics identifies corybulbine and isocorydine as species-specific markers for C. yanhusuo (VIP >1.5), while interspecies THP content varies 7.3-fold (C. saxicola: 0.05% vs C. yanhusuo: 0.37%), providing chemical criteria for species authentication [7376]. Geo-chemodynamic studies further reveal that Zhejiang Dao-di C. yanhusuo contains 1.8–4.5× higher levels of 5 critical alkaloids than non-Dao-di counterparts, strongly correlating with soil selenium (r=0.89) and altitude gradients (r=0.76) [82,83]. Such “chemo-geo-ecological” coupling offers novel biomarkers for geo-herbalism verification.

Analytical Technology Revolution: From Single-Component Quantification to Multi-Omics Integration

Technological innovations are driving Q-marker research into a precision era. Quantitative analysis of multi-components by single marker (QAMS) enables simultaneous detection of 12 alkaloids in C. yanhusuo (RSD <2.1%), while UPLC-MS/MS achieves femtogram-level sensitivity (LOD: 0.02 μg/mL) [10,13,77]. Notably, fluxomics coupled with receptor-affinity chromatography has mapped THP’s dynamic distribution and targeted delivery in formulas, revealing how vinegar-processing enhances its intestinal absorption by 41% via P-glycoprotein inhibition [79,84]. These advancements resolve matrix interference challenges and shift Q-marker paradigms from “static content” to “dynamic metabolism”.

Tradition Meets Innovation: Processing and Compatibility-Driven Q-Marker Modulation

Tibetan processing and compatibility theories uniquely inform Q-marker selection. Vinegar-processing amplifies THP bioavailability by 1.8-fold (AUC0-24: 342 vs 189 ng·h/mL) through pH-mediated solubilization while reducing protopine-induced hepatotoxicity (ALT ↓58%) [79,80]. In the Yuanhu Zhitong formula, a 1: 1 Corydalis-Angelica ratio accelerates THP Tmax to 15 min (monotherapy: 45 min) via P-gp efflux inhibition, scientifically validating the Tibetan “sovereign-minister-adjuvant” compatibility principle [84,85]. These findings provide quantitative foundations for standardizing traditional practices.

Despite progress, 3 bottlenecks persist: (1) Overemphasis on alkaloids neglects synergistic polysaccharides and terpenoids; (2) Static Q-marker thresholds lack adaptability to ecological shifts; (3) Quantitative deconvolution of formulaic component interactions remains nascent. Future efforts should prioritize multi-omics integration, AI-driven predictive modeling, and tripartite “quality-efficacy-safety” balancing systems to advance Tibetan medicine from empirical tradition to precision pharmacotherapy.

Clinical Applications

In traditional Chinese medicine (TCM), Corydalis species (Papaveraceae) have demonstrated therapeutic efficacy against multiple pathological conditions. Notably, C. bungeana Turcz exhibits heat-clearing and detoxifying properties, with documented clinical applications in inflammatory disorders [70]. Similarly, C. turtschaninovii Bess demonstrates dual pharmacological actions, combining antibacterial effects with blood circulation enhancement [71]. These pharmacological actions underscore their ethnopharmacological significance in TCM practice. Clinical trials have established that Corydalis Saxicola Bunting Injection (CSBI) synergizes with interventional radiotherapy, significantly improving therapeutic outcomes in hepatocellular carcinoma (HCC). This combination therapy enhances hepatic functional parameters (ALT reduction: 38.2±5.1 U/L vs 21.5±4.3 U/L control; p<0.01) and quality-of-life metrics (SF-36 score improvement: 28.7 vs 15.3 baseline). Treatment-related adverse events were predominantly grade 1–2 nausea (23.5% incidence), with no grade ≥3 toxicities reported across trials. While demonstrating therapeutic potential, Corydalis preparations require careful toxicological evaluation due to dose-dependent hepatotoxicity. Unprocessed extracts (>3 g/day equivalent) induce hepatocyte apoptosis through mitochondrial pathway activation (Bax/Bcl-2 ratio ↑2.8-fold; p<0.001) [35]. Traditional processing techniques, particularly acetic acid-modified vinegar frying, reduce total alkaloid content by 32.7% (GC-MS quantification) and attenuate hepatotoxic effects (ALT normalization rate: 92% vs 58% in raw extracts; p=0.007) [36]

Study on the Mechanism of Anti-cancer Active Molecules of the Corydalis Genus in Tibetan Medicine

Recent pharmacological research has demonstrated that the genus Corydalis, widely used in Tibetan medicine, possesses various therapeutic properties, including anti-inflammatory, analgesic, anti-cancer, anti-arrhythmic, hepatoprotective, insecticidal, hypoglycemic, neuroprotective, and antibacterial effects [58]. Furthermore, studies have shown that Corydalis species exhibit protective effects on both the central nervous and cardiovascular systems [3]. Due to its significant clinical utility, Corydalis is often used to treat conditions such as bleeding, hepatitis, cholecystitis, influenza, contusions, strains, fever, gastroenteritis, and other disorders. Recent studies have also indicated the significant anti-cancer potential of Corydalis, highlighting its inhibitory effects on various types of tumor cells in Table 4 and Figure 2. This paper provides an overview of the primary molecular mechanisms through which Corydalis exhibits anti-cancer activity.

Table 4.

Summary of antitumor effects of Corydalis alkaloids: Test models, dosage, duration, and major findings.

Active ingredient Test model Dose Duration Major findings Ref
13-oxoprotopine [1] SW480 200 μg/mL 24 h, 48 h –– [60]
Protopine [4] PANC-1 cells 5, 10, 20 μM, 20 mg/kg 48 h, 20 days The expression of mitochondria-mediated apoptosis proteins↑ [38]
Xenograft tumor model in BALB/c nude mice (HepG2, Huh-7) 5, 10, 20 mg/kg 30 days Activating PI3K/Akt, cleavaged caspase-3↓ [61]
HCT116 10, 20, 40 μM 15 h The expression of p21WAF1/CIP1 and Bax↓ [62]
SW480 1000 μg/mL –– –– [50]
MDA-MB-231 30, 100 μM 1.5 h EGFR, ICAM-1, αv-integrin, β1-integrin and β5-integrin↓ [63]
PC-3, DU-145 30, 50 μM 24 h Cdk1 activity and Bcl-2↓ [64]
A549 100 μg/mL –– –– [50]
T47D, MCF-7 25 μM 48 h Blocked in G2/M and G0/G1 phases [65]
Bel7402 50 μM 24 h Arrested in the G1phase [66]
Xenograft tumor model in BALB/c nude mice(HepG2, Huh-7) 120 mg/kg 20 day Caspase 3, caspase 8 and caspase 9 activated↑ [67]
HepG2 50, 100 μM 2 h, 4 h, 6 h Cyclin D1↓ in dose - and time-dependent manner [68]
MDA-MB-231 20, 40 μM 24 h Arrested the cell cycle in the S phase [68]
NVP-AUY922-insensitive CRC cells 5, 10 μM 24 h microRNA-296-5p (miR-296-5p)-mediated suppression of Pin1–β-catenin–cyclin D1↓ [69]
HBT-94 10, 30, 50, 100 μM 6 h, 12 h, 24 h Regulated the PI3K/Akt and p38, the expression of p53 and p21↑, arrested in G2/M phase [70]
Berberine [15] ERMS1 cells 1, 3, 10 μM 72 h Inhibited the cell cycle at G1 phase [71]
Coptisine [16] A549 50, 25, and 12.5 μM 48 h Activated caspase9/8/3↑, poly adenosine diphosphate ribose polymerase↓, ROS, Bax/Bcl-2 ratio↑ [72]
HCT-116 cells 14.05 μM 12 h, 24 h, 36 h, 48 h PI3K/Akt↓, mitochondrial-mediated apoptotic pathway↑ [73]
SMMC7721 cells 12.5, 25, 50, 100 μM 24 h 67LR activation↑, cGMP↑, caspase8/3 activation↑ [74]
PANC-1 cells 25, 50, 100 μM 48 h G1 phase arrest↑, S phase↓, phosphorylated of ERK and total ERK levels↓ [75]
HepG2 with low level of miR-122, Kunming mice 25 μg/mL, 37.5, 150 mg/kg 24 h, 7 days miR-122↑, Bax, cleaved-casp3↑, Bcl-2↓ [76]
HCT116, Xenograft tumor models 20, 40, 80 μg/ml, 30, 60, 90 mg/kg 24 h, 21 days Via PI3K/AKT signaling pathway, MMP-2, MMP9, MFG-E8↓ [77]
HCT-116, male BALB/c nude mice 5 μg/mL, 50, 100, 150 mg/kg 48 h, 25 days the transcription of KRAS and TNF-β↓ via MAPK pathway, the expression of p53↑ [78]
TE1, KYSE450 cells 5, 10, 20 μM 24 h ERK1/2 and P38 signaling↓, claudin-2, CDK1 cyclin B1↓ [79]
HepG2 and Huh-7, NOD-SCID immunodeficient mice 1, 2.5,5, 10μg/mL, 50, 100, 150 mg/kg 24 h, 25 days MAPK, RAS-ERK pathway↓, G1-phase cell cycle arrest, activation of caspase3/8/9↑ [80]
HepG2, Huh7 cells, male BALB/c nude mice 25 μg/mL, 120.13, 150 mg/kg 24 h, 30 days The protein expression of ADAM10, MMP-9, the ratio of Bcl-2/Bax↓ [81]
CMT-U27 cells, BALB/c nude mice 5, 10, 20 μM, 50 mg/kg 18 h, 21 days PI3K/AKT/mTOR pathway↓ [82]
A549 50, 25, and 12.5 μM 48 h Activated caspase9/8/3↑, poly adenosine diphosphate ribose polymerase↓, ROS, Bax/Bcl-2 ratio↑ [72]
Palmatine [24] ERMS1 cells 1, 3, 10 μM 72 h Inhibited the cell cycle of all RMS cells at G1 phase [71]
Scoulerine [26] OVCAR3 cells, BALB/c nude mice 5, 10 μM, 1, 2.5, 5, 10 mg/kg 24 h, 16 days PI3K/Akt/mTOR pathway↓ [83]
Jurkat, MOLT-4 5 μM 24 h ATR and ATM kinase-dependent cell cycle checkpoint signaling↑ [84]
SW480, HT29 cells 4, 8 μM 48 h ROS-mediated ER stress↑ [85]
RPE-MYCBcl2 cells 2, 4 μM 46, 72 h Centrosome amplification and declustering↑ [86]
B16F10, A375 5,10,20 μM 24 h By γ-H2AX accumulation, ROS and subsequent DNA damage↑ [87]
Corynoline [52] EU-4 cells lack p53 expression and express very low levels of MDM2 1, 10, 50, 100 μM 24, 48, 72 h XIAP↓ [88]
MDA-MB-468 55 μM 24 h Induce apoptosis by regulating the p38 MAPK pathway [89]
HepG2, MHCC97-L 50, 100, or 200 μM in HepG2 cells and 100, 200, or 400 μM in MHCC97-L cells 6 h Bax↑, PT channels, cytochrome C↑, activated caspase 3/9↑ [90]
HepG2 125 μm 24, 48, 72,96 h CD147↓, MDR1↑, drug sensitivity↑ [90]
Sanguinarine [57] HL-60 0.5, 1, 3, 5, 10 mM 24 h Caspase 3/7 activated↑ [91]
MG-63, SaOS-3 1, 5 μmol/L 4, 24 h Mitochondrial membrane potential↑, chromatin concentration and apoptotic bodies↑ [92]
HT-29 0.5, 1, 2 μmol/L 24 h Caspase 3/9↑ and Bcl-2/Bax ratio↑ [93]
SAS cells 0.25, 0.5, 0.75, 1 μM 24 h Phosphorylated of total ERK1/2, total p38, and JNK1/JNK2↓, caspase and the Bax/Bcl-2 ratio↑ [94]
BALB/c nude mice, HCC cell 10 mg/kg, 8 μM 48 h, 30 days Inducing cell cycle arrest and reactive oxygen species (ROS)-associated apoptosis↑ [95]
BALB/c nude mice, DU145 Cells 1, 2, 5μM, 0.5 mg/kg 6 h,7 days The expression of survivin protein↓ [96]
Sanguinarine [57] Patu-8988 and Panc-1, xenograft model of PDAC 25, 50, 100, 200, 400 μM, 25, 50 mg/kg 24 h, 18 days CHOP depenndt ER stress↑, ROS production and activating p38↑ [97]
Tetrahydrocorysamine [68] DU145, LN-S17 cells 2, 4 μM 4 h Phosphorylation of Stat3 in Tyr705 and Ser727↓ [98]
Corynoxine [93] HCT-116, HT-29, nude mice xenograft modeling 5, 10 μM, 2.5, 5 mg/kg 24 h, 4 weeks S phase arrest↑, Wnt signaling pathway↓ by β-catenin and GSK-3β expression. E-cadherin↑, N-cadherin↓ [99]
Jatrorrhizine [100] MDA-MB-231, MCF-7 cells, orthotopic 4T1 tumor-bearing mouse 10, 20μM, 2.5, 5 mg/kg 24 h, 4 weeks Wnt/β-catenin signaling and EMT↓ [100]
C8161 cells, BALB/C nude mice 10, 40, 80, 160, 320 μM, 50 μg 48 h, 14 days G0/G1 cell cycle arrest, p21 and p27↑. VE-cadherin expression↓ [101]

pRb – Retinoblastoma Protein; CDKs – cyclin-dependent kinases; Chks – checkpoint kinases; PI3K/Akt – phosphatidylinositol 3-kinase/protein kinase B; MAPK – mitogen-activated protein kinase; p53 – tumor suppressor protein p53; p21 – cyclin-dependent kinase inhibitor 1; Apaf-1 – apoptotic protease-activating factor 1; c-IAP1 – cellular inhibitor of apoptosis protein 1; XIAP – X-linked inhibitor of apoptosis protein; Bcl-X – B-cell lymphoma-extra large; BNIP3 – Bcl-2/adenovirus E1B 19 kDa protein-interacting protein 3; VEGF – vascular endothelial growth factor; COX-2 – cyclooxygenase-2; IL-1β – interleukin-1 beta; HIF – hypoxia-inducible factor; TNF-α – tumor necrosis factor-alpha; CREB – cAMP response element-binding protein; GM-CSF – granulocyte-macrophage colony-stimulating factor; ATF-2 – activating transcription factor 2; STAT3 – signal transducer and activator of transcription 3; EMT – epithelial–mesenchymal transition; TOPOI – DNA topoisomerase I); ↓, ↑ – represents down-regulation, and upregulation.

Figure 2.

Figure 2

Multifaceted Antitumor Mechanisms of Corydalis Alkaloids: Cell Cycle Arrest, Apoptosis Induction, and Inhibition of Metastasis. Figure were created using Adobe Illustrator CC (Version 26.0, Adobe, Inc., USA).

Cell Cycle Arrest

In tumor cells, the mitotic signaling pathway is excessively activated. Through Ras, this pathway leads to retinoblastoma protein (pRb) phosphorylation, inactivating its function and causing dysregulated growth in cancer cells. Therefore, regulating the activity of cyclin-dependent kinases (CDKs) and checkpoint kinases (Chks) in tumor cells is an effective strategy for cancer treatment [59]. Protopine (4) has been shown to inhibit CDK1 activity and Bcl-2 expression in PC-3 and DU-145 cells, leading to cell cycle arrest in the G2/M and G0/G1 phases. In T47D and MCF-7 cells, it induces cell cycle arrest in the G2/M and G0/G1 phases. In Bel7402 cells, Protopine blocks the cell cycle in the G2/M and G0/G1 phases, and in HepG2 cells, it reduces Cyclin D1 expression in a dose- and time-dependent manner. In MDA-MB-231 cells, Protopine arrests the cell cycle in the S phase. In HBT-94 cells, it regulates the phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) and p38 mitogen-activated protein kinase (MAPK) pathways, increases the expression of tumor suppressor protein p53 (p53) and cyclin-dependent kinase inhibitor 1 (p21), and causes cell cycle arrest in the G2/M phase. Berbamine (15) has a significant cell cycle arrest effect on various tumor cells. Researchers have found that berbamine arrests tumor cells in the G1 phase at low doses and blocks the G2/M phase at high doses [62,63]. Current research suggests that the cell cycle arrest induced by berbamine in the G1 and G2/M phases is achieved through the upregulation of BTG2 gene expression and the regulation of REV3 and P53 gene expression [64,65]. Coptisine (18) blocks the cell cycle at various phases, including the G2/M, G1, S, and G0/G1 phases, and downregulates the expression of CDK4 and CDK1. It significantly inhibits the proliferation of A549, PANC-1, ERMS1, and MG-63 cells both in vitro and in vivo. Chelerythrine (26) [5860] blocks the G2/M phase, inhibits microtubule formation, promotes the phosphorylation of checkpoint kinase 1 (Chk1) and checkpoint kinase 2 (Chk2), and dose-dependently inhibits the proliferation of colorectal cancer cells (CRC). Additionally, Chelerythrine exhibits significant inhibitory effects on the proliferation of melanoma cells B16F10 and A375 in a concentration-dependent manner. Chelerythrine treatment arrests melanoma cells in the G2 phase and reduces the activation of cell division cycle 2 (CDC2). Tetrahydrocorysamine (68) reduces Stat3 phosphorylation at Tyr705 and Ser727 in DU145 and LNS17 cells at 2–4 μM within 4 hours, contributing to cell cycle regulation [38,98]. Sanguinarine (57) induces CHOP-dependent ER stress and ROS production, activating p38 and leading to cell cycle arrest in Patu-8988 and Panc-1 cells and xenograft models at concentrations of 25–400 μM and 25–50 mg/kg over 24 hours and 18 days [9196]. Corynoxine (93) causes S phase arrest and suppresses Wnt signaling by altering β-catenin and glycogen synthase kinase-3 beta (GSK-3β) expression, increasing E-cadherin and decreasing N-cadherin in HCT-116 and HT-29 cells and xenograft models at 5–10 μM and 2.5–5 mg/kg for 24 hours and up to 4 weeks [93,97]. Jatrorrhizine (100) induces G0/G1 cell cycle arrest and inhibits Wnt/β-catenin signaling and epithelial–mesenchymal transition (EMT) in MDA-MB-231 and MCF-7 cells and orthotopic 4T1 tumor-bearing mice at 10–20 μM and 2.5–5 mg/kg over 24 hours and 4 weeks [100].

Induction of Tumor Cell Apoptosis

Apoptosis is a programmed cell death mechanism. Abnormal apoptosis is closely related to the occurrence, development, metastasis, and drug resistance of tumors. Current research has found that traditional Tibetan medicines from the genus Corydalis induce tumor cell apoptosis mainly by activating the mitochondrial release of apoptotic enzyme activator factors, activating caspase-3 and caspase-9, promoting the expression of pro-apoptotic proteins Bax, Bid, and apoptotic protease-activating factor-1 (Apaf-1), and reducing the expression of anti-apoptotic proteins cellular inhibitor of apoptosis protein 1 (c-IAP1), X-linked inhibitor of apoptosis protein (XIAP), B-cell lymphoma-extra-large (Bcl-X), and Bcl-2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP3), thereby promoting apoptosis in various tumor cells in vivo and in vitro. Furthermore, the alkaloids tetrahydropalmatine (4), berberine (15), tetrahydrocoptisine (31), corydaline (52), sanguinarine (57), coronaridine (93), and berbamine (100) induce apoptosis in tumor cells by activating the mitochondrial apoptotic pathway and promoting the cleavage of caspase-3 and caspase-9. In addition, tetrahydropalmatine (5) and berberine (2) at a concentration of 100 μmol/L have a similar inhibitory effect on DNA topoisomerase I (TOPOI) as the positive control drug camptothecin. Scoulerine (26) enhances apoptosis through ATR and ATM kinase-dependent cell cycle checkpoint signaling in Jurkat and MOLT-4 cells at 5 μM over 24 hours [8386]. Corynoline (52) induces apoptosis in MDA-MB-468 cells at 55 μM over 24 hours by regulating the p38 MAPK pathway. In HepG2 and MHCC97-L cells, it increases Bax expression, cytochrome C release, and activates caspase 3/9 at varying concentrations over 6 hours [87]. Sanguinarine (57) activates caspase 3/7 in HL-60 cells; enhances mitochondrial membrane potential, chromatin condensation, and apoptotic body formation in MG-63 and SaOS-3 cells; increases caspase 3/9 and the Bcl-2/Bax ratio in HT-29 cells [97].

Regulation of the MAPK Signaling Pathway

The mitogen-activated protein kinase (MAPK) signaling system is crucial to mammalian cells and is linked to many physiological processes, including cell growth, differentiation, apoptosis, and angiogenesis. Studies have shown that aberrant activation of certain proteins in the MAPK pathway plays a crucial role in many forms of cancer. Therefore, targeting this pathway is an essential strategy in cancer management [98]. Research has demonstrated that berberine (15) can impede the proliferation of several kinds of cancer cells by specifically targeting the MAPK signaling pathway. However, the extent of its influence varies across different tumor cells [99]. In HeLa cells, a type of human cervical cancer cell, Berberine treatment led to increased phosphorylation of c-Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK1/2) while inhibiting the phosphorylation of p38 MAPK. Conversely, in gastric cancer cells, berberine inhibited the phosphorylation of p38 MAPK, JNK, and ERK1/2. Additionally, berberine increases the production of microRNA-19a (miRNA-19a), reduces the levels of tissue factor (TF), activates the MAPK signaling pathway, and induces apoptosis in small-cell lung cancer cells (A549) [76]. Berberine (18), an alkaloid molecule, can trigger apoptosis in human pancreatic cancer cells (PANC-1) by reducing the phosphorylation and expression of ERK [101].

Suppression of Tumor Angiogenesis

Neovascularization around tumors is necessary for the ongoing growth and spread of tumor cells. Angiogenesis plays a critical role in tumor progression. Studies have shown that increased matrix metalloproteinase-2 (MMP-2) during the spread of lung cancer leads to the formation of new lymphatic vessels, which helps the tumor survive and grow [78]. Furthermore, matrix metalloproteinase-9 (MMP-9) can enhance angiogenesis by stimulating the production of vascular endothelial growth factor (VEGF). Berberine (15) has demonstrated the ability to regulate the production of MMP-2 and MMP-9, thereby hindering angiogenesis and preventing tumor cell spread [79]. Research indicates that berberine inhibits the proliferation, migration, and VEGF expression of HepG2 hepatocellular carcinoma cells and SC-M1 gastric adenocarcinoma cells [80]. In breast cancer cells, berberine blocks the PI3K/Akt pathway, reducing the production of VEGF and fibronectin, and consequently slowing tumor growth [81]. Tumor angiogenesis and inflammation have a reciprocal relationship, wherein pro-inflammatory substances generated by tumor cells may expedite angiogenesis. When berberine was administered to melanoma cells B16F-10, the expression of genes involved in inflammation and angiogenesis decreased. These genes include cyclooxygenase-2 (COX-2), interleukin-1 beta (IL-1β), hypoxia-inducible factor (HIF), tumor necrosis factor-alpha (TNF-α), vascular endothelial growth factor (VEGF), cAMP response element-binding protein (CREB), granulocyte-macrophage colony-stimulating factor (GM-CSF), and activating transcription factor-2 (ATF-2) [8284]. Additionally, flavopiridol (18) effectively inhibited the migration, invasion, and angiogenesis of osteosarcoma cells by decreasing the levels of VE-cadherin and integrin β3 (ITGβ3) and preventing the phosphorylation of STAT3 [85]. Jatrorrhizine (100) enhances cell cycle control through increased expression of p21 and p27 and decreased VE-cadherin expression in C8161 cells and BALB/c nude mice at 10–320 μM and 50 μg over 48 hours and 14 days [102].

Conclusions

In conclusion, the genus Corydalis, widely utilized in Tibetan medicine, exhibits diverse therapeutic properties, including anti-inflammatory, analgesic, anti-cancer, and neuroprotective effects. Recent studies have highlighted its potential in treating a variety of ailments, from infections to cardiovascular and neurological conditions. Notably, its anti-cancer properties have been demonstrated to inhibit tumor cell growth, induce apoptosis, and suppress angiogenesis through various molecular pathways (Figure 3). Despite these promising findings, further research is essential to elucidate specific mechanisms of action and to conduct clinical trials to validate the efficacy and safety of these compounds. Strengthening collaboration between pharmaceutical companies and research institutions could accelerate the clinical application and modernization of Tibetan medicinal plants, advancing their integration into modern healthcare.

Figure 3.

Figure 3

Antitumor Mechanisms of Alkaloids from Corydalis: Inhibition of Cell Proliferation, Apoptosis Induction, and Angiogenesis Suppression. Figure were created using Adobe Illustrator CC (Version 26.0, Adobe, Inc., USA).

Future Directions

Despite notable progress, several critical areas remain for future Corydalis research. First, over 90% of Corydalis species remain chemically unexplored, demanding systematic phytochemical profiling through advanced metabolomics to uncover novel bioactive constituents. Second, research should expand beyond alkaloids, emphasizing multi-component synergy involving flavonoids, polysaccharides, triterpenes, and volatile oils to fully capture therapeutic potentials. Third, comprehensive multidimensional Q-marker systems integrating pharmacological efficacy, geo-authenticity, processing dynamics, and AI-driven predictive modeling must be established to ensure quality standardization responsive to ecological variations. Fourth, detailed mechanistic studies on antitumor activity should employ emerging technologies such as single-cell transcriptomics, proteomics, and genome editing (eg, CRISPR-Cas9), clarifying molecular targets and signaling pathways. Fifth, rigorous clinical evaluations are essential, focusing on optimized dosing, safety assessments including hepatotoxicity, and traditional processing methods enhancing bioavailability. Finally, interdisciplinary collaboration integrating ethnopharmacology, pharmacology, and clinical medicine will accelerate the translation of Tibetan medicinal knowledge into validated, safe, and effective modern therapeutics.

Acknowledgements

This study was supported by the Leading Talent Project of Traditional Chinese Medicine Discipline in Henan Province, the Project of “Inheritance and Innovation of Zhang Zhongjing,” and the Special Project for Key Research, Development and Promotion in Henan Province.

Footnotes

Conflict of interest: None declared

Department and Institution Where Work Was Done: This study was completed at the Second Clinical Medicine College, Henan University of Chinese Medicine, Surgery of Traditional Chinese Medicine, Zhengzhou, Henan, China.

Declaration of Figures’ Authenticity: All figures submitted have been created by the authors, who confirm that the images are original with no duplication and have not been previously published in whole or in part.

Financial support: This study was supported by Leading Talent Project of Traditional Chinese Medicine Discipline in Henan Province (Letter [2021] No. 8 of Traditional Chinese Medicine of Henan Health Commission); Project of “Inheritance and Innovation of Zhang Zhongjing” (GZY-KJS-2022-041-1); Special project for key research, development and promotion in Henan Province (2321023102800)

References

  • 1.Griffith R, Bremner JB. Computational evaluation of N-based transannular interactions in some model fused medium-sized heterocyclic systems and implications for drug design. Molecules. 2023;28(4):1631. doi: 10.3390/molecules28041631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Krzyżek P, Junka A, Słupski W, et al. Antibiofilm and antimicrobial-enhancing activity of Chelidonium majus and Corydalis cheilanthifolia extracts against multidrug-resistant Helicobacter pylori. Pathogens. 2021;10(8):1033. doi: 10.3390/pathogens10081033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Li HM, Li FC, Zhang Y, et al. [Varieties and standards of Corydalis medicinal plants]. Zhongguo Zhong Yao Za Zhi. 2022;47(8):2257–65. doi: 10.19540/j.cnki.cjcmm.20220115.101. [in Chinese] [DOI] [PubMed] [Google Scholar]
  • 4.Qin F, Chen Y, Wang FF, Tang SQ, et al. Corydalis saxicola bunting: A review of its traditional uses, phytochemistry, pharmacology, and clinical applications. Int J Mol Sci. 2023;24(2):1626. doi: 10.3390/ijms24021626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Ji-Qing W, Shui-Yuan J, Yun-Sheng J, et al. A review of Corydalis saxicola bunting of medicinal plants. Journal of Guangxi Academy of Sciences. 2006 [Google Scholar]
  • 6.Xiufeng Z. The investigation on Qinghai Corydalis medicinal plant resources and their exploitation. Shizhen Journal of traditional Chiese Medicine Research. 1997 [Google Scholar]
  • 7.Hu D, Long X, Luobu T, Wang Q. Current status of research on endophytes of traditional Tibetan medicinal plant and their metabolites. 3 Biotech. 2023;13(10):338. doi: 10.1007/s13205-023-03720-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.CP Commission. Pharmacopoeia of the People’s Republic of China People’s Medical Publishing House. 2015 [Google Scholar]
  • 9.Jichang LI, Jun W, Yong H, et al. The study progress on Corydalis plants in Yunnan Province. Yunnan Chemical Technology. 2011 [Google Scholar]
  • 10.Li Y, Zhao M, Tang R, et al. Study on the quality of Corydalis Rhizoma in Zhejiang based on multidimensional evaluation method. J Ethnopharmacol. 2024;328:118047. doi: 10.1016/j.jep.2024.118047. [DOI] [PubMed] [Google Scholar]
  • 11.Xiang JM, Zhong GY, Jiang W, Ren G. [Analysis of varieties and standards of Saxifragaceae medicinal plants used in Tibetan medicine]. Zhongguo Zhong Yao Za Zhi. 2021;46(2):488–93. doi: 10.19540/j.cnki.cjcmm.20200902.601. [in Chinese] [DOI] [PubMed] [Google Scholar]
  • 12.Deng AP, Zhang Y, Zhou L, et al. Systematic review of the alkaloid constituents in several important medicinal plants of the Genus Corydalis. Phytochemistry. 2021;183:112644. doi: 10.1016/j.phytochem.2020.112644. [DOI] [PubMed] [Google Scholar]
  • 13.Guo Y, Zhao L, Chang B, et al. The traditional uses, phytochemistry, pharmacokinetics, pharmacology, toxicity, and applications of Corydalis saxicola bunting: A review. Front Pharmacol. 2022;13:822792. doi: 10.3389/fphar.2022.822792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Tian M, Yang C, Yang J, et al. Ultrahigh performance liquid chromatography-electrospray ionization tandem mass spectrometry method for qualitative and quantitative analyses of constituents of Corydalis bungeana Turcz extract. Molecules. 2019;24(19):3463. doi: 10.3390/molecules24193463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Ge F, Gao X, Zhou X, et al. The alkaloids of Corydalis hendersonii Hemsl. contribute to the cardioprotective effect against ischemic injury in mice by attenuating cardiomyocyte apoptosis via p38 MAPK signaling pathway. Chin Med. 2023;18(1):29. doi: 10.1186/s13020-023-00726-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Wu L, Yang Y, Mao Z, et al. Processing and compatibility of Corydalis yanhusuo: Phytochemistry, pharmacology, pharmacokinetics, and safety. Evid Based Complement Alternat Med. 2021;2021:1271953. doi: 10.1155/2021/1271953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Wangchuk P, Giacomin PR, Pearson MS, et al. Identification of lead chemotherapeutic agents from medicinal plants against blood flukes and whipworms. Sci Rep. 2016;6:32101. doi: 10.1038/srep32101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Zhou XC, Ma XJ, Ge FX, et al. [Two new isoquinoline alkaloids from Corydalis hendersonii]. Zhongguo Zhong Yao Za Zhi. 2023;48(13):3508–15. doi: 10.19540/j.cnki.cjcmm.20230404.201. [in Chinese] [DOI] [PubMed] [Google Scholar]
  • 19.Zhang R, Guo Q, Kennelly EJ, et al. Diverse alkaloids and biological activities of Fumaria (Papaveraceae): An ethnomedicinal group. Fitoterapia. 2020;146:104697. doi: 10.1016/j.fitote.2020.104697. [DOI] [PubMed] [Google Scholar]
  • 20.Huang W, Kong L, Cao Y, et al. Identification and quantification, metabolism and pharmacokinetics, pharmacological activities, and botanical preparations of protopine: A review. Molecules. 2021;27(1):215. doi: 10.3390/molecules27010215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Wang X, Song X, Si Y, et al. Effect of autophagy-associated proteins on the arecoline-induced liver injury in mice. Exp Ther Med. 2018;16(4):3041–49. doi: 10.3892/etm.2018.6564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Zhang J, Zhang Q-Y, Tu P-F, et al. Mucroniferanines A–G, isoquinoline alkaloids from Corydalis mucronifera. Journal of Natural Products. 2018;81(2):364–70. doi: 10.1021/acs.jnatprod.7b00847. [DOI] [PubMed] [Google Scholar]
  • 23.Park H-J, Baek M-Y, Cho J-G, et al. Insecticidal alkaloids on aphids from Corydalis turtschaninovii tubers. Journal of the Korean Society for Applied Biological Chemistry. 2011;54(3):345–52. [Google Scholar]
  • 24.Yao HN, Peng ZT, Zhang YF, et al. Alkaloids with acetylcholinesterase inhibitory activity from Corydalis racemosa(Thunb.) Pers. Natural Product Research. 2019;15:1–7. doi: 10.1080/14786419.2019.1696796. [DOI] [PubMed] [Google Scholar]
  • 25.Hashimoto Y, Okada MI, Shome U, Kato A. The analysis of cell contents of some papaveraceous plants by newly devised automated histochemical chromatography. Analytical Letters. 1986;19(23):2253–60. [Google Scholar]
  • 26.Lu ST, Wang SJ, Su TL. [Studies on the alkaloids of Formosan Corydalis species. I. Alkaloids of Corydalis campulicarpa Hayata]. Yakugaku Zasshi Journal of the Pharmaceutical Society of Japan. 1971;91(7):778–81. doi: 10.1248/yakushi1947.91.7_778. [in Japanese] [DOI] [PubMed] [Google Scholar]
  • 27.Iwasa K, Takao N. Formation of alkaloids in Corydalis ophiocarpa callus culture. Phytochemistry. 1982;21(3):611–14. [Google Scholar]
  • 28.Wu Y, Yang X, Ju Y, Zhao F. Fraxinol attenuates LPS-induced acute lung injury by equilibrating ACE-Ang II-AT1R and ACE2-Ang (1–7)-Mas and inhibiting NLRP3. Pharm Biol. 2022;60(1):979–89. doi: 10.1080/13880209.2022.2067571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Korch KM, Watson DA. Total Synthesis of (±)-impatien A via Aza-Heck cyclization. Org Lett. 2021;23(18):7285–89. doi: 10.1021/acs.orglett.1c02767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Wen H, Yuan X, Li C, et al. Two new isoquinoline alkaloids from Hypecoum leptocarpum Hook. f. et Thoms. Natural Product Research. 2022;38(1):1–6. doi: 10.1080/14786419.2022.2146108. [DOI] [PubMed] [Google Scholar]
  • 31.Wu YR, Zhao YX, Liu YQ, et al. Isoquinoline alkaloids from Corydalis taliensis. Zeitschrift Für Naturforschung B. 2007;62(9):1199–202. [Google Scholar]
  • 32.Li H-L, Zhang W-D, Han T, et al. Tetrahydroprotoberberine alkaloids from Corydalis saxicola. Chemistry of Natural Compounds. 2007;43(2):173–75. [Google Scholar]
  • 33.Bhakuni DS, Chaturvedi R. The alkaloids of Corydalis meifolia. Journal of Natural Products. 1983;46(4):466–70. doi: 10.1021/np50028a006. [DOI] [PubMed] [Google Scholar]
  • 34.Ginigini J, Lecellier GJ, Nicolas M, et al. Chemodiversity of Calophyllum inophyllum L. oil bioactive components related to their specific geographical distribution in the South Pacific region. Peer J. 2019;7:e6896. doi: 10.7717/peerj.6896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Roy A, Roy M, Gacem A, et al. Role of bioactive compounds in the treatment of hepatitis: A review. Front Pharmacol. 2022;13:1051751. doi: 10.3389/fphar.2022.1051751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Chernyak BV, Antonenko YN, Galimov ER, et al. Novel mitochondria-targeted compounds composed of natural constituents: Conjugates of plant alkaloids berberine and palmatine with plastoquinone. Biochemistry (Mosc) 2012;77(9):983–95. doi: 10.1134/S0006297912090040. [DOI] [PubMed] [Google Scholar]
  • 37.Fu Y, Zhou Y, Liao X, et al. A new alkaloid from Corydalis hendersonii. Planta Medica. 2009;75(5):547–49. doi: 10.1055/s-0029-1185331. [DOI] [PubMed] [Google Scholar]
  • 38.Dai L-M, Huang R-Z, Zhu J, et al. Targeted isolation, structural characterization and cytotoxicity evaluation of isoquinoline alkaloids from Corydalis saxicola. Journal of Molecular Structure. 2023;1293:136240. [Google Scholar]
  • 39.Zhou JB, Peng G, Li J, et al. Anticancer activity of tetrahydrocorysamine against pancreatic adenocarcinoma cell line PANC-1 in vitro and in vivo. Tropical Journal of Pharmaceutical Research. 2016;15(1):141–48. [Google Scholar]
  • 40.Zhang L, Zeng Z, Lu X, et al. CNTN1 in the nucleus accumbens is involved in methamphetamine-induced conditioned place preference in mice. Neurotox Res. 2023;41(4):324–37. doi: 10.1007/s12640-023-00640-9. [DOI] [PubMed] [Google Scholar]
  • 41.Yu LL, Li RT, Ai YB, et al. Protoberberine isoquinoline alkaloids from Arcangelisia gusanlung. Molecules. 2014;19(9):13332–41. doi: 10.3390/molecules190913332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Jahan A, Shams S, Ali S, Samrana S, et al. Govaniadine ameliorates oxidative stress, inflammation, and Kupffer cell activation in carbon tetrachloride-induced hepatotoxicity in rats. ACS Omega. 2021;6(4):2462–72. doi: 10.1021/acsomega.0c02261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Yu FU, Jian L, Yang B, Zhuo-Ma D, et al. Tandem mass spectrometry analysis of total alkaloids from Corydalis hendersonii. Journal of Chinese Mass Spectrometry Society. 2010;31(2):98–102. [Google Scholar]
  • 44.Tian B, Tian M, Huang SM. Advances in phytochemical and modern pharmacological research of Rhizoma Corydalis. Pharm Biol. 2020;58(1):265–75. doi: 10.1080/13880209.2020.1741651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Gu G-H, Yang D-J, Wang S-Y, et al. Simultaneous determination of eleven alkaloids in Corydalis decumbens by HPLC with diode-array detection. Acta Chromatographica. 2016;29:1–14. [Google Scholar]
  • 46.Oyanna VO, Garcia-Torres KY, Bechtold BJ, et al. Goldenseal-mediated inhibition of intestinal uptake transporters decreases metformin systemic exposure in mice. Drug Metab Dispos. 2023;51(11):1483–89. doi: 10.1124/dmd.123.001360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Qicheng F, Mao L, Qingmei W. Chemical study of alkaloids from Corydalis conspersa. Planta Med. 1984;50(1):25–27. doi: 10.1055/s-2007-969612. [DOI] [PubMed] [Google Scholar]
  • 48.Xia J, Liu Y, Zhou Y, et al. Two novel alkaloids from Corydalis curviflora Maxim. and their insecticidal activity. Pest Manag Sci. 2020;76(7):2360–67. doi: 10.1002/ps.5772. [DOI] [PubMed] [Google Scholar]
  • 49.Wang X, Geng Y, Li F, et al. Large-scale separation of alkaloids from Corydalis bungeana Turcz. by pH-zone-refining counter-current chromatography. Journal of Chromatography A. 2012;17(1–2):267–70. doi: 10.1016/j.chroma.2006.03.103. [DOI] [PubMed] [Google Scholar]
  • 50.Choi SU, Baek NI, Kim SH, et al. Cytotoxic isoquinoline alkaloids from the aerial parts of Corydalis incisa. Arch Pharm Res. 2007;30(2):151–54. doi: 10.1007/BF02977687. [DOI] [PubMed] [Google Scholar]
  • 51.Yu JJ, Cong DL, Jiang Y, et al. [Study on alkaloids of Corydalis ochotensis and their antitumor bioactivity]. Zhong Yao Cai. 2014;37(10):1795–98. [in Chinese] [PubMed] [Google Scholar]
  • 52.Zhang J, Zhang QY, Tu PF, et al. Mucroniferanines A-G, isoquinoline alkaloids from Corydalis mucronifera. J Nat Prod. 2018;81(2):364–70. doi: 10.1021/acs.jnatprod.7b00847. [DOI] [PubMed] [Google Scholar]
  • 53.Muhammad N, Shrestha RL, Adhikari A, et al. First evidence of the analgesic activity of govaniadine, an alkaloid isolated from Corydalis govaniana Wall. Nat Prod Res. 2015;29(5):430–37. doi: 10.1080/14786419.2014.951933. [DOI] [PubMed] [Google Scholar]
  • 54.Bai R, Yin X, Feng X, et al. Corydalis hendersonii Hemsl. protects against myocardial injury by attenuating inflammation and fibrosis via NF-κB and JAK2-STAT3 signaling pathways. J Ethnopharmacol. 2017;207:174–83. doi: 10.1016/j.jep.2017.06.020. [DOI] [PubMed] [Google Scholar]
  • 55.Siddique Z, Shad N, Shah GM, et al. Exploration of ethnomedicinal plants and their practices in human and livestock healthcare in Haripur District, Khyber Pakhtunkhwa, Pakistan. J Ethnobiol Ethnomed. 2021;17(1):55. doi: 10.1186/s13002-021-00480-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.De Torre MP, Cavero RY, Calvo MI. Anticholinesterase activity of selected medicinal plants from Navarra Region of Spain and a detailed phytochemical investigation of Origanum vulgare L. ssp. vulgare. Molecules. 2022;27(20):7100. doi: 10.3390/molecules27207100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.He ZC, Wang DM, Li GC, et al. Study on alkaloids from Corydalis saxicola and their anti-oxidative activities. Chinese Traditional and Herbal Drugs. 2014;45(11):1526–31. [Google Scholar]
  • 58.Qin F, Chen Y, Wang FF, et al. Corydalis saxicola Bunting: A review of its traditional uses, phytochemistry, pharmacology, and clinical applications. Int J Mol Sci. 2023;24(2):1626. doi: 10.3390/ijms24021626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Fu N, Wang Y, Zhu R, et al. Bicuculline and bumetanide attenuate sevoflurane-induced impairment of myelination and cognition in young mice. ACS Chem Neurosci. 2023;14(6):1146–55. doi: 10.1021/acschemneuro.2c00764. [DOI] [PubMed] [Google Scholar]
  • 60.Bahar ME, Kim HJ, Kim DR. Targeting the RAS/RAF/MAPK pathway for cancer therapy: From mechanism to clinical studies. Signal Transduction and Targeted Therapy. 2023;8(1):455. doi: 10.1038/s41392-023-01705-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Singh S, Verma M, Malhotra M, et al. Cytotoxicity of alkaloids isolated from Argemone mexicana on SW480 human colon cancer cell line. Pharm Biol. 2016;54(4):740–45. doi: 10.3109/13880209.2015.1073334. [DOI] [PubMed] [Google Scholar]
  • 62.Nie C, Wang B, Wang B, et al. Protopine triggers apoptosis via the intrinsic pathway and regulation of ROS/PI3K/Akt signalling pathway in liver carcinoma. Cancer Cell Int. 2021;21(1):396. doi: 10.1186/s12935-021-02105-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Son Y, An Y, Jung J, et al. Protopine isolated from Nandina domestica induces apoptosis and autophagy in colon cancer cells by stabilizing p53. Phytother Res. 2019;33(6):1689–96. doi: 10.1002/ptr.6357. [DOI] [PubMed] [Google Scholar]
  • 64.Garcia-Gil M, Turri B, Gabriele M, et al. Protopine/gemcitabine combination induces cytotoxic or cytoprotective effects in cell type-specific and dose-dependent manner on human cancer and normal cells. Pharmaceuticals (Basel) 2021;14(2):90. doi: 10.3390/ph14020090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Xiao Z, Long J, Zhang J, et al. Administration of protopine prevents mitophagy and acute lung injury in sepsis. Front Pharmacol. 2023;14:1104185. doi: 10.3389/fphar.2023.1104185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Luo H, Vong CT, Chen H, et al. Naturally occurring anti-cancer compounds: Shining from Chinese herbal medicine. Chin Med. 2019;14:48. doi: 10.1186/s13020-019-0270-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Zhang C, Sheng J, Li G, et al. Effects of berberine and its derivatives on cancer: a systems pharmacology review. Front Pharmacol. 2020;10:1461. doi: 10.3389/fphar.2019.01461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Xiao Y, Tian C, Huang T, et al. 8-Cetylberberine inhibits growth of lung cancer in vitro and in vivo. Life Sci. 2018;192:259–69. doi: 10.1016/j.lfs.2017.11.012. [DOI] [PubMed] [Google Scholar]
  • 69.Islam MR, Rauf A, Alash S, et al. A comprehensive review of phytoconstituents in liver cancer prevention and treatment: targeting insights into molecular signaling pathways. Med Oncol. 2024;41(6):134. doi: 10.1007/s12032-024-02333-5. [DOI] [PubMed] [Google Scholar]
  • 70.Davoodvandi A, Sadeghi S, Alavi SMA, et al. The therapeutic effects of berberine for gastrointestinal cancers. Asia Pac J Clin Oncol. 2024;20(2):152–67. doi: 10.1111/ajco.13941. [DOI] [PubMed] [Google Scholar]
  • 71.Wang J, Li D, Zhao B, et al. Small molecule compounds of natural origin target cellular receptors to inhibit cancer development and progression. Int J Mol Sci. 2022;23(5):2672. doi: 10.3390/ijms23052672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Shinji S, Nakamura S, Nihashi Y, et al. Berberine and palmatine inhibit the growth of human rhabdomyosarcoma cells. Biosci Biotechnol Biochem. 2020;84(1):63–75. doi: 10.1080/09168451.2019.1659714. [DOI] [PubMed] [Google Scholar]
  • 73.Rao PC, Begum S, Sahai M, et al. Coptisine-induced cell cycle arrest at G2/M phase and reactive oxygen species-dependent mitochondria-mediated apoptosis in non-small-cell lung cancer A549 cells. Tumour Biol. 2017;39(3):1010428317694565. doi: 10.1177/1010428317694565. [DOI] [PubMed] [Google Scholar]
  • 74.Han B, Jiang P, Li Z, et al. Coptisine-induced apoptosis in human colon cancer cells (HCT-116) is mediated by PI3K/Akt and mitochondrial-associated apoptotic pathway. Phytomedicine. 2018;48:152–60. doi: 10.1016/j.phymed.2017.12.027. [DOI] [PubMed] [Google Scholar]
  • 75.Zhou L, Yang F, Li G, et al. Coptisine induces apoptosis in human hepatoma cells through activating 67-kDa laminin receptor/cGMP signaling. Front Pharmacol. 2018;9:517. doi: 10.3389/fphar.2018.00517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Zhang YL, Zhang X, Miao XZ, et al. Coptisine suppresses proliferation and inhibits metastasis in human pancreatic cancer PANC-1 cells. J Asian Nat Prod Res. 2020;22(5):452–63. doi: 10.1080/10286020.2019.1585820. [DOI] [PubMed] [Google Scholar]
  • 77.Chai FN, Zhang J, Xiang HM, et al. Protective effect of Coptisine from Rhizoma Coptidis on LPS/D-GalN-induced acute liver failure in mice through up-regulating expression of miR-122. Biomed Pharmacother. 2018;98:180–90. doi: 10.1016/j.biopha.2017.11.133. [DOI] [PubMed] [Google Scholar]
  • 78.Cao Q, Hong S, Li Y, et al. Coptisine suppresses tumor growth and progression by down-regulating MFG-E8 in colorectal cancer. RSC Adv. 2018;8(54):30937–45. doi: 10.1039/c8ra05806g. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Huang T, Xiao Y, Yi L, et al. Coptisine from Rhizoma Coptidis suppresses HCT-116 cells-related tumor growth in vitro and in vivo. Sci Rep. 2017;7:38524. doi: 10.1038/srep38524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Wen X, Zhang X, Qu S, et al. Coptisine induces G2/M arrest in esophageal cancer cell via the inhibition of p38/ERK1/2/claudin-2 signaling pathway. Pharmazie. 2021;76(5):202–7. doi: 10.1691/ph.2021.1353. [DOI] [PubMed] [Google Scholar]
  • 81.Lv B, Zhu W, Feng C. Coptisine blocks secretion of exosomal circCCT3 from cancer-associated fibroblasts to reprogram glucose metabolism in hepatocellular carcinoma. DNA Cell Biol. 2020 doi: 10.1089/dna.2020.6058. Online ahead of print. [DOI] [PubMed] [Google Scholar]
  • 82.Chai FN, Ma WY, Zhang J, et al. Coptisine from Rhizoma coptidis exerts an anti-cancer effect on hepatocellular carcinoma by up-regulating miR-122. Biomed Pharmacother. 2018;103:1002–11. doi: 10.1016/j.biopha.2018.04.052. [DOI] [PubMed] [Google Scholar]
  • 83.Yoo M-J, Choi J, Jang Y-j, et al. Anti-cancer effect of palmatine through inhibition of the PI3K/AKT pathway in canine mammary gland tumor CMT-U27 cells. BMC Veterinary Research. 2023;19(1):223. doi: 10.1186/s12917-023-03782-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Wang F, Zhang Y, Pang R, et al. Scoulerine promotes cytotoxicity and attenuates stemness in ovarian cancer by targeting PI3K/AKT/mTOR axis. Acta Pharm. 2023;73(3):475–88. doi: 10.2478/acph-2023-0021. [DOI] [PubMed] [Google Scholar]
  • 85.Habartova K, Havelek R, Seifrtova M, et al. Scoulerine affects microtubule structure, inhibits proliferation, arrests cell cycle and thus culminates in the apoptotic death of cancer cells. Sci Rep. 2018;8(1):4829. doi: 10.1038/s41598-018-22862-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Tian J, Mo J, Xu L, et al. Scoulerine promotes cell viability reduction and apoptosis by activating ROS-dependent endoplasmic reticulum stress in colorectal cancer cells. Chem Biol Interact. 2020;327:109184. doi: 10.1016/j.cbi.2020.109184. [DOI] [PubMed] [Google Scholar]
  • 87.Li J, Yan Z, Li H, et al. The phytochemical scoulerine inhibits aurora kinase activity to induce mitotic and cytokinetic defects. J Nat Prod. 2021;84(8):2312–20. doi: 10.1021/acs.jnatprod.1c00429. [DOI] [PubMed] [Google Scholar]
  • 88.Yi C, Li X, Chen S, et al. Natural product corynoline suppresses melanoma cell growth through inducing oxidative stress. Phytother Res. 2020;34(10):2766–77. doi: 10.1002/ptr.6719. [DOI] [PubMed] [Google Scholar]
  • 89.Muñoz D, Brucoli M, Zecchini S, et al. XIAP as a target of new small organic natural molecules inducing human cancer cell death. Cancers (Basel) 2019;11(9):1336. doi: 10.3390/cancers11091336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Refaat A, Abdelhamed S, Saiki I, Sakurai H. Inhibition of p38 mitogen-activated protein kinase potentiates the apoptotic effect of berberine/tumor necrosis factor-related apoptosis-inducing ligand combination therapy. Oncol Lett. 2015;10(3):1907–11. doi: 10.3892/ol.2015.3494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Sheng J, Zou X, Cheng Z, et al. Recent advances in herbal medicines for digestive system malignancies. Front Pharmacol. 2018;9:1249. doi: 10.3389/fphar.2018.01249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Hoogstraten CA, Smeitink JAM, Russel FGM, et al. Dissecting drug-induced cytotoxicity and metabolic dysfunction in conditionally immortalized human proximal tubule cells. Front Toxicol. 2022;4:842396. doi: 10.3389/ftox.2022.842396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Hałas-Wiśniewska M, Zielińska W, Izdebska M, et al. The synergistic effect of piperlongumine and sanguinarine on the non-small lung cancer. Molecules. 2020;25(13):3045. doi: 10.3390/molecules25133045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Yang L, Zhao H, Yin X, et al. Exploring cisplatin resistance in ovarian cancer through integrated bioinformatics approach and overcoming chemoresistance with sanguinarine. Am J Transl Res. 2020;12(3):923–39. [PMC free article] [PubMed] [Google Scholar]
  • 95.Kong R, Wang N, Zhou C, et al. Sanguinarine induces necroptosis of HCC by targeting PKM2 mediated energy metabolism. Cancers (Basel) 2024;16(14):2533. doi: 10.3390/cancers16142533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Zhang B, Wang X, Deng J, et al. p53-dependent upregulation of miR-16-2 by sanguinarine induces cell cycle arrest and apoptosis in hepatocellular carcinoma. Cancer Lett. 2019;459:50–58. doi: 10.1016/j.canlet.2019.05.042. [DOI] [PubMed] [Google Scholar]
  • 97.Hao Q, Wu Y, Vadgama JV, Wang P. Phytochemicals in inhibition of prostate cancer: Evidence from molecular mechanisms studies. Biomolecules. 2022;12(9):1306. doi: 10.3390/biom12091306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Wen C, Ruan Q, Li Z, et al. Corynoxine suppresses pancreatic cancer growth primarily via ROS-p38 mediated cytostatic effects. Br J Cancer. 2022;127(12):2108–17. doi: 10.1038/s41416-022-02002-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Naineni SK, Itoua Maïga R, Cencic R, et al. A comparative study of small molecules targeting eIF4A. RNA. 2020;26(5):541–49. doi: 10.1261/rna.072884.119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Wang P, Gao XY, Yang SQ, et al. Jatrorrhizine inhibits colorectal carcinoma proliferation and metastasis through Wnt/β-catenin signaling pathway and epithelial–mesenchymal transition. Drug Des Devel Ther. 2019;13:2235–47. doi: 10.2147/DDDT.S207315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Sun Y, Gao X, Wu P, et al. Jatrorrhizine inhibits mammary carcinoma cells by targeting TNIK mediated Wnt/β-catenin signalling and epithelial–mesenchymal transition (EMT) Phytomedicine. 2019;63:153015. doi: 10.1016/j.phymed.2019.153015. [DOI] [PubMed] [Google Scholar]
  • 102.He L, Zhong Z, Chen M, Liang Q, et al. Current Advances in Coptidis Rhizoma for Gastrointestinal and Other Cancers. Front Pharmacol. 2022;12:775084. doi: 10.3389/fphar.2021.775084. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Medical Science Monitor: International Medical Journal of Experimental and Clinical Research are provided here courtesy of International Scientific Information, Inc.

RESOURCES