A Systematic Literature Review of Breast Cancer Diagnosis Using Machine Intelligence Techniques

  • Review article
  • Published: 11 April 2022
  • Volume 29 , pages 4401–4430, ( 2022 )

Cite this article

breast cancer a systematic literature review

  • Varsha Nemade 1 ,
  • Sunil Pathak 1 &
  • Ashutosh Kumar Dubey 2  

1895 Accesses

21 Citations

Explore all metrics

Breast cancer is one of the most common diseases in women; it can have long-term implications and can even be fatal. However, early detection, achieved through recent advancements in technology, can help reduce mortality. In this paper, different machine intelligence techniques [machine learning (ML), and deep learning (DL)] were analysed in the context of breast cancer. In addition, the classification of breast cancer into malignant and benign using different breast cancer image modalities were discussed. Furthermore, the diagnosis of breast cancer using various publicly and privately available image datasets, pre-processing techniques, feature extraction techniques, comparison between conventional ML and different convolutional neural network (CNN) architectures, and transfer learning techniques were discussed in detail. It also correlates the parameters and attributes impact in case of different methods applied. Advantages and the limitations of the machine intelligence approaches were highlighted based on the discussion and analysis. A total of 162 research publications was considered for the time period of 2015–2021. These are in the chronological order of their appearance. This systematic literature review will be helpful to the researchers due to the detailed analysis of different methodologies and in conducting further investigations.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Subscribe and save.

  • Get 10 units per month
  • Download Article/Chapter or eBook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime

Price includes VAT (Russian Federation)

Instant access to the full article PDF.

Rent this article via DeepDyve

Institutional subscriptions

breast cancer a systematic literature review

Similar content being viewed by others

breast cancer a systematic literature review

Deep learning-based breast cancer classification through medical imaging modalities: state of the art and research challenges

breast cancer a systematic literature review

Comparative Analysis of Classification and Detection of Breast Cancer from Histopathology Images Using Deep Neural Network

breast cancer a systematic literature review

Recent Advancement and Challenges of Deep Learning for Breast Mass Classification from Mammogram Images

Explore related subjects.

  • Artificial Intelligence
  • Medical Imaging

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F (2021) Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. Cancer J Clin 71(3):209–249

Article   Google Scholar  

Ferlay J, Colombet M, Soerjomataram I, Parkin DM, Piñeros M, Znaor A, Bray F (2021) Cancer statistics for the year 2020: an overview. Int J Cancer 149:778–789

Li J, Zhou Z, Dong J, Fu Y, Li Y, Luan Z, Peng X (2021) Predicting breast cancer 5-year survival using machine learning: A systematic review. PLoS ONE 16(4):e0250370

Freeman K, Geppert J, Stinton C, Todkill D, Johnson S, Clarke A, Taylor-Phillips S (2021) Use of artificial intelligence for image analysis in breast cancer screening programmes: systematic review of test accuracy. BMJ. https://doi.org/10.1136/bmj.n1872

Dubey AK, Gupta U, Jain S (2016) Epidemiology of lung cancer and approaches for its prediction: a systematic review and analysis. Chin J Cancer 35(1):1–3

Dubey AK, Gupta U, Jain S (2015) Breast cancer statistics and prediction methodology: a systematic review and analysis. Asian Pac J Cancer Prev 16(10):4237–4245

Ashhar SM, Mokri SS, Abd Rahni AA, Huddin AB, Zulkarnain N, Azmi NA, Mahaletchumy T (2021) Comparison of deep learning convolutional neural network (CNN) architectures for CT lung cancer classification. Int J Adv Technol Eng Explor 8(74):126

Guo Q, Wang X, Gao Y, Zhou J, Huang C, Zhang Z, Chu H (2021) Relationship between particulate matter exposure and female breast cancer incidence and mortality: a systematic review and meta-analysis. Int Arch Occup Environ Health 94(2):191–201

Siegel RL, Miller KD, Jemal A (2015) Cancer statistics, 2015. Cancer J Clin 65(1):5–29

Dubey AK, Gupta U, Jain S (2016) Analysis of k-means clustering approach on the breast cancer Wisconsin dataset. Int J Comput Assist Radiol Surg 11(11):2033–2047

Iranmakani S, Mortezazadeh T, Sajadian F, Ghaziani MF, Ghafari A, Khezerloo D, Musa AE (2020) A review of various modalities in breast imaging: technical aspects and clinical outcomes. Egypt J Radiol Nucl Med 51:1–22

Kumar R, Srivastava R, Srivastava S (2015) Detection and classification of cancer from microscopic biopsy images using clinically significant and biologically interpretable features. J Med Eng. https://doi.org/10.1155/2015/457906

Zhou X, Li C, Rahaman MM, Yao Y, Ai S, Sun C, Wang Q, Zhang Y, Li M, Li X, Jiang T (2020) A comprehensive review for breast histopathology image analysis using classical and deep neural networks. IEEE Access 8:90931–90956

Tsochatzidis L, Koutla P, Costaridou L, Pratikakis I (2021) Integrating segmentation information into CNN for breast cancer diagnosis of mammographic masses. Comput Methods Prog Biomed 200:105913

Abdelhafiz D, Yang C, Ammar R, Nabavi S (2019) Deep convolutional neural networks for mammography: advances, challenges and applications. BMC Bioinf 20(11):1–20

Google Scholar  

Al-Antari MA, Al-Masni MA, Choi MT, Han SM, Kim TS (2018) A fully integrated computer-aided diagnosis system for digital X-ray mammograms via deep learning detection, segmentation, and classification. Int J Med Inf 117:44–54

Al-Antari MA, Han SM, Kim TS (2020) Evaluation of deep learning detection and classification towards computer-aided diagnosis of breast lesions in digital X-ray mammograms. Comput Methods Prog Biomed. 196:105584

Gnanasekaran VS, Joypaul S, Sundaram PM, Chairman DD (2020) Deep learning algorithm for breast masses classification in mammograms. IET Image Proc 14(12):2860–2868

Nagarajan V, Britto EC, Veeraputhiran SM (2019) Feature extraction based on empirical mode decomposition for automatic mass classification of mammogram images. Med Novel Technol Dev. 1:100004

Peng W, Mayorga RV, Hussein EM (2016) An automated confirmatory system for analysis of mammograms. Comput Methods Programs Biomed 125:134–144

Al-Masni MA, Al-Antari MA, Park JM, Gi G, Kim TY, Rivera P, Valarezo E, Choi MT, Han SM, Kim TS (2018) Simultaneous detection and classification of breast masses in digital mammograms via a deep learning YOLO-based CAD system. Comput Methods Programs Biomed 157:85–94

Zheng J, Lin D, Gao Z, Wang S, He M, Fan J (2020) Deep learning assisted efficient AdaBoost algorithm for breast cancer detection and early diagnosis. IEEE Access 8:96946–96954

Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gøtzsche PC, Ioannidis JP, Clarke M, Devereaux PJ, Kleijnen J, Moher D (2009) The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration. J Clin Epidemiol 62(10):e1-34

Russakovsky O, Deng J, Su H, Krause J, Satheesh S, Ma S, Huang Z, Karpathy A, Khosla A, Bernstein M, Berg AC (2015) Imagenet large scale visual recognition challenge. Int J Comput Vision 115(3):211–252

Article   MathSciNet   Google Scholar  

Guan S, Loew M (2017) Breast cancer detection using transfer learning in convolutional neural networks. In: 2017 IEEE applied imagery pattern recognition workshop (AIPR). IEEE, pp 1–8

Seemendra A, Singh R, Singh S (2021) Breast cancer classification using transfer learning. In: Evolving technologies for computing, communication and smart world 2021. Springer, Singapore, pp 425–436

Kumar Y, Gupta S, Singla R, Hu YC (2021) A systematic review of artificial intelligence techniques in cancer prediction and diagnosis. Arch Comput Methods Eng. https://doi.org/10.1007/s11831-021-09648-w

Abdelhafiz D, Bi J, Ammar R, Yang C, Nabavi S (2020) Convolutional neural network for automated mass segmentation in mammography. BMC Bioinf 21(1):1–9

Hossain MS (2019) Microc alcification segmentation using modified U-net segmentation network from mammogram images. J King Saud Univ Comput Inf Sci. 34:86–94

Chougrad H, Zouaki H, Alheyane O (2020) Multi-label transfer learning for the early diagnosis of breast cancer. Neurocomputing 392:168–180

Wang Z, Wang S, Zhu Y, Ma Y (2016) Review of image fusion based on pulse-coupled neural network. Arch Comput Methods Eng 23(4):659–671

Article   MathSciNet   MATH   Google Scholar  

Tiong LC, Kim ST, Ro YM (2019) Implementation of multimodal biometric recognition via multi-feature deep learning networks and feature fusion. Multimed Tools Appl 78(16):22743–22772

Singh S, Mittal N, Singh H (2021) Review of various image fusion algorithms and image fusion performance metric. Arch Comput Methods Eng. 28:3645–3659

Dubey AK, Gupta U, Jain S (2022) Medical data clustering and classification using TLBO and machine learning algorithms. CMC-Comput Mater Continua 70(3):4523–4543

Dhahri H, Al Maghayreh E, Mahmood A, Elkilani W, Faisal Nagi M (2019) Automated breast cancer diagnosis based on machine learning algorithms. J Healthcare Eng. https://doi.org/10.1155/2019/4253641

Mohammed SA, Darrab S, Noaman SA, Saake G (2020) Analysis of breast cancer detection using different machine learning techniques. In: International conference on data mining and big data. Springer, Singapore, pp 108–117

Lomboy KE, Hernandez RM (2021) A comparative performance of breast cancer classification using hyper-parameterized machine learning models. Int J Adv Technol Eng Explor 8(82):1080–1101

Melekoodappattu JG, Subbian PS (2020) Automated breast cancer detection using hybrid extreme learning machine classifier. J Ambient Intell Human Comput. https://doi.org/10.1007/s12652-020-02359-3

Buda M, Saha A, Walsh R, Ghate S, Li N, Święcicki A, Lo JY, Mazurowski MA (2021) A data set and deep learning algorithm for the detection of masses and architectural distortions in digital breast tomosynthesis images. JAMA Netw Open 4(8):e2119100

Chugh G, Kumar S, Singh N (2021) Survey on machine learning and deep learning applications in breast cancer diagnosis. Cogn Comput 13:1451–1470

Dhivya P, Bazilabanu A, Ponniah T (2021) Machine learning model for breast cancer data analysis using triplet feature selection algorithm. IETE J Res. https://doi.org/10.1080/03772063.2021.1963861

Singh OV, Choudhary P, Thongam K (2019) A study on deep learning for breast cancer detection in histopathological images. In: International conference on computer vision and image processing 2019. Springer, Singapore, pp 36–48

Priyanka KS (2021) A review paper on breast cancer detection using deep learning. In: IOP conference series: materials science and engineering 2021, vol 1022, no 1. IOP Publishing, p 012071

Ma L, Lu G, Wang D, Qin X, Chen ZG, Fei B (2019) Adaptive deep learning for head and neck cancer detection using hyperspectral imaging. Visual Comput Ind Biomed Art 2(1):1–2

Nayak DR, Dash R, Majhi B, Pachori RB, Zhang Y (2020) A deep stacked random vector functional link network autoencoder for diagnosis of brain abnormalities and breast cancer. Biomed Signal Process Control 58:101860

Rahman AS, Belhaouari SB, Bouzerdoum A, Baali H, Alam T, Eldaraa AM (2020) Breast mass tumor classification using deep learning. In: 2020 IEEE international conference on informatics, IoT, and enabling technologies (ICIoT). IEEE, pp 271–276

Alkhaleefah M, Chittem PK, Achhannagari VP, Ma SC, Chang YL (2020) The influence of image augmentation on breast lesion classification using transfer learning. In: 2020 International conference on artificial intelligence and signal processing (AISP). IEEE, pp 1–5

Khan HN, Shahid AR, Raza B, Dar AH, Alquhayz H (2019) Multi-view feature fusion based four views model for mammogram classification using convolutional neural network. IEEE Access 7:165724–165733

Lu HC, Loh EW, Huang SC (2019) The classification of mammogram using convolutional neural network with specific image preprocessing for breast cancer detection. In: 2019 International conference on artificial intelligence and big data. IEEE, pp 9–12

Chougrad H, Zouaki H, Alheyane O (2018) Deep convolutional neural networks for breast cancer screening. Comput Methods Programs Biomed 157:19–30

Yemini M, Zigel Y, Lederman D (2018) Detecting masses in mammograms using convolutional neural networks and transfer learning. In: 2018 IEEE international conference on the science of electrical engineering in Israel (ICSEE). IEEE, pp 1–4

Yu S, Liu L, Wang Z, Dai G, Xie Y (2019) Transferring deep neural networks for the differentiation of mammographic breast lesions. Sci China Technol Sci 62(3):441–447

Zhang X, Zhang Y, Han EY, Jacobs N, Han Q, Wang X, Liu J (2018) Classification of whole mammogram and tomosynthesis images using deep convolutional neural networks. IEEE Trans Nanobiosci 17(3):237–242

Arevalo J, González FA, Ramos-Pollán R, Oliveira JL, Lopez MA (2016) Representation learning for mammography mass lesion classification with convolutional neural networks. Comput Methods Programs Biomed 127:248–257

Jadoon MM, Zhang Q, Haq IU, Butt S, Jadoon A (2017) Three-class mammogram classification based on descriptive CNN features. BioMed Res Int. https://doi.org/10.1155/2017/3640901

Al-Najdawi N, Biltawi M, Tedmori S (2015) Mammogram image visual enhancement, mass segmentation and classification. Appl Soft Comput 35:175–185

Gomathi P, Muniraj C, Periasamy PS (2020) Breast thermography based unsupervised anisotropic-feature transformation method for automatic breast cancer detection. Microprocess Microsyst 77:103137

Jen CC, Yu SS (2015) Automatic detection of abnormal mammograms in mammographic images. Expert Syst Appl 42(6):3048–3055

Carvalho ED, Antonio Filho OC, Silva RR, Araujo FH, Diniz JO, Silva AC, Paiva AC, Gattass M (2020) Breast cancer diagnosis from histopathological images using textural features and CBIR. Artifi Intell Med. 105:101845

Song R, Li T, Wang Y (2020) Mammographic classification based on XGBoost and DCNN with multi features. IEEE Access 8:75011–75021

Shen L, He M, Shen N, Yousefi N, Wang C, Liu G (2020) Optimal breast tumor diagnosis using discrete wavelet transform and deep belief network based on improved sunflower optimization method. Biomed Signal Process Control 60:101953

Abdel-Nasser M, Melendez J, Moreno A, Omer OA, Puig D (2017) Breast tumor classification in ultrasound images using texture analysis and super-resolution methods. Eng Appl Artif Intell 59:84–89

Muramatsu C, Hara T, Endo T, Fujita H (2016) Breast mass classification on mammograms using radial local ternary patterns. Comput Biol Med 72:43–53

da Rocha SV, Junior GB, Silva AC, de Paiva AC, Gattass M (2016) Texture analysis of masses malignant in mammograms images using a combined approach of diversity index and local binary patterns distribution. Expert Syst Appl 66:7–19

Nahid AA, Kong Y (2018) Histopathological breast-image classification using local and frequency domains by convolutional neural network. Information 9(1):19

Bruno DO, Do Nascimento MZ, Ramos RP, Batista VR, Neves LA, Martins AS (2016) LBP operators on curvelet coefficients as an algorithm to describe texture in breast cancer tissues. Expert Syst Appl 55:329–340

Muduli D, Dash R, Majhi B (2020) Automated breast cancer detection in digital mammograms: a moth flame optimization based ELM approach. Biomed Signal Process Control 59:101912

Sun W, Tseng TL, Zhang J, Qian W (2017) Enhancing deep convolutional neural network scheme for breast cancer diagnosis with unlabeled data. Comput Med Imaging Graph 57:4–9

Magna G, Casti P, Jayaraman SV, Salmeri M, Mencattini A, Martinelli E, Di Natale C (2016) Identification of mammography anomalies for breast cancer detection by an ensemble of classification models based on artificial immune system. Knowl-Based Syst 101:60–70

Kaur P, Singh G, Kaur P (2019) Intellectual detection and validation of automated mammogram breast cancer images by multi-class SVM using deep learning classification. Inf Med Unlock. 16:100151

Raghavendra U, Acharya UR, Fujita H, Gudigar A, Tan JH, Chokkadi S (2016) Application of Gabor wavelet and Locality Sensitive Discriminant Analysis for automated identification of breast cancer using digitized mammogram images. Appl Soft Comput 46:151–161

Sannasi Chakravarthy SR, Rajaguru H (2019) Comparison analysis of linear discriminant analysis and cuckoo-search algorithm in the classification of breast cancer from digital mammograms. Asian Pac J Cancer Prev 20(8):2333

Mohanty F, Rup S, Dash B (2020) Automated diagnosis of breast cancer using parameter optimized kernel extreme learning machine. Biomed Signal Process Control 62:102108

Mohanty F, Rup S, Dash B, Majhi B, Swamy MN (2020) An improved scheme for digital mammogram classification using weighted chaotic salp swarm algorithm-based kernel extreme learning machine. Appl Soft Comput 91:106266

Bejnordi BE, Mullooly M, Pfeiffer RM, Fan S, Vacek PM, Weaver DL, Herschorn S, Brinton LA, van Ginneken B, Karssemeijer N, Beck AH (2018) Using deep convolutional neural networks to identify and classify tumor-associated stroma in diagnostic breast biopsies. Mod Pathol 31(10):1502–1512

de Oliveira FS, de Carvalho Filho AO, Silva AC, de Paiva AC, Gattass M (2015) Classification of breast regions as mass and non-mass based on digital mammograms using taxonomic indexes and SVM. Comput Biol Med 57:42–53

Miranda GH, Felipe JC (2015) Computer-aided diagnosis system based on fuzzy logic for breast cancer categorization. Comput Biol Med 64:334–346

He W, Hogg P, Juette A, Denton ER, Zwiggelaar R (2015) Breast image pre-processing for mammographic tissue segmentation. Comput Biol Med 67:61–73

Li Y, Chen H, Yang Y, Cheng L, Cao L (2015) A bilateral analysis scheme for false positive reduction in mammogram mass detection. Comput Biol Med 57:84–95

Khan S, Hussain M, Aboalsamh H, Bebis G (2017) A comparison of different Gabor feature extraction approaches for mass classification in mammography. Multimed Tools Appl 76(1):33–57

Huynh BQ, Li H, Giger ML (2016) Digital mammographic tumor classification using transfer learning from deep convolutional neural networks. J Med Imaging 3(3):034501

Kallenberg M, Petersen K, Nielsen M, Ng AY, Diao P, Igel C, Vachon CM, Holland K, Winkel RR, Karssemeijer N, Lillholm M (2016) Unsupervised deep learning applied to breast density segmentation and mammographic risk scoring. IEEE Trans Med Imaging 35(5):1322–1331

Li Y, Chen H, Wei X, Peng Y, Cheng L (2016) Mass classification in mammograms based on two-concentric masks and discriminating texton. Pattern Recogn 60:648–656

Wang Z, Qu Q, Yu G, Kang Y (2016) Breast tumor detection in double views mammography based on extreme learning machine. Neural Comput Appl 27(1):227–240

Swiderski B, Osowski S, Kurek J, Kruk M, Lugowska I, Rutkowski P, Barhoumi W (2017) Novel methods of image description and ensemble of classifiers in application to mammogram analysis. Expert Syst Appl 81:67–78

Cordeiro FR, Santos WP, Silva-Filho AG (2017) Analysis of supervised and semi-supervised GrowCut applied to segmentation of masses in mammography images. Comput Methods Biomech Biomed Eng: Imaging Visual 5(4):297–315

Casti P, Mencattini A, Salmeri M, Ancona A, Lorusso M, Pepe ML, Di Natale C, Martinelli E (2017) Towards localization of malignant sites of asymmetry across bilateral mammograms. Comput Methods Programs Biomed 140:11–18

Carneiro G, Nascimento J, Bradley AP (2017) Automated analysis of unregistered multi-view mammograms with deep learning. IEEE Trans Med Imaging 36(11):2355–2365

Byra M, Dobruch-Sobczak K, Klimonda Z, Piotrzkowska-Wroblewska H, Litniewski J (2020) Early prediction of response to neoadjuvant chemotherapy in breast cancer sonography using Siamese convolutional neural networks. IEEE J Biomed Health Inf 25:797

Dhungel N, Carneiro G, Bradley AP (2017) A deep learning approach for the analysis of masses in mammograms with minimal user intervention. Med Image Anal 37:114–128

Bardou D, Zhang K, Ahmad SM (2018) Classification of breast cancer based on histology images using convolutional neural networks. Ieee Access 6:24680–24693

Ribli D, Horváth A, Unger Z, Pollner P, Csabai I (2018) Detecting and classifying lesions in mammograms with deep learning. Sci Rep 8(1):1–7

Vo DM, Nguyen NQ, Lee SW (2019) Classification of breast cancer histology images using incremental boosting convolution networks. Inf Sci 482:123–138

Ting FF, Tan YJ, Sim KS (2019) Convolutional neural network improvement for breast cancer classification. Expert Syst Appl 120:103–115

Cai H, Huang Q, Rong W, Song Y, Li J, Wang J, Chen J, Li L (2019) Breast microcalcification diagnosis using deep convolutional neural network from digital mammograms. Comput Math Methods Med. https://doi.org/10.1155/2019/2717454

Article   MATH   Google Scholar  

Li H, Zhuang S, Li DA, Zhao J, Ma Y (2019) Benign and malignant classification of mammogram images based on deep learning. Biomed Signal Process Control 51:347–354

Herent P, Schmauch B, Jehanno P, Dehaene O, Saillard C, Balleyguier C, Arfi-Rouche J, Jégou S (2019) Detection and characterization of MRI breast lesions using deep learning. Diagn Interv Imaging 100(4):219–225

Vijayarajeswari R, Parthasarathy P, Vivekanandan S, Basha AA (2019) Classification of mammogram for early detection of breast cancer using SVM classifier and Hough transform. Measurement 146:800–805

Bevilacqua V, Brunetti A, Guerriero A, Trotta GF, Telegrafo M, Moschetta M (2019) A performance comparison between shallow and deeper neural networks supervised classification of tomosynthesis breast lesions images. Cogn Syst Res 53:3–19

Fang Y, Zhao J, Hu L, Ying X, Pan Y, Wang X (2019) Image classification toward breast cancer using deeply-learned quality features. J Visual Commun Image Represent 64:102609

Li Y, Wu J, Wu Q (2019) Classification of breast cancer histology images using multi-size and discriminative patches based on deep learning. IEEE Access 7:21400–21408

Zhu Z, Harowicz M, Zhang J, Saha A, Grimm LJ, Hwang ES, Mazurowski MA (2019) Deep learning analysis of breast MRIs for prediction of occult invasive disease in ductal carcinoma in situ. Comput Biol Med 115:103498

Wang Z, Li M, Wang H, Jiang H, Yao Y, Zhang H, Xin J (2019) Breast cancer detection using extreme learning machine based on feature fusion with CNN deep features. IEEE Access 7:105146–105158

Kumar A, Singh SK, Saxena S, Lakshmanan K, Sangaiah AK, Chauhan H, Shrivastava S, Singh RK (2020) Deep feature learning for histopathological image classification of canine mammary tumors and human breast cancer. Inf Sci 508:405–421

Vaka AR, Soni B, Reddy S (2020) Breast cancer detection by leveraging Machine Learning. ICT Express 6(4):320–324

Dabass J, Hanmandlu M, Vig R (2020) Classification of digital mammograms using information set features and Hanman Transform based classifiers. Inf Med Unlock. 20:100401

Agarwal R, Díaz O, Yap MH, Lladó X, Martí R (2020) Deep learning for mass detection in Full Field Digital Mammograms. Comput Biol Med 121:103774

Shen T, Wang J, Gou C, Wang FY (2020) Hierarchical fused model with deep learning and type-2 fuzzy learning for breast cancer diagnosis. IEEE Trans Fuzzy Syst 28(12):3204–3218

Chouhan N, Khan A, Shah JZ, Hussnain M, Khan MW (2021) Deep convolutional neural network and emotional learning based breast cancer detection using digital mammography. Comput Biol Med 132:104318

Shen T, Hao K, Gou C, Wang FY (2021) Mass Image Synthesis in Mammogram with Contextual Information Based on GANs. Comput Methods Program Biomed. 202:106019

Yan Y, Conze PH, Lamard M, Quellec G, Cochener B, Coatrieux G (2021) Towards improved breast mass detection using dual-view mammogram matching. Med Image Anal 71:102083

Soulami KB, Kaabouch N, Saidi MN, Tamtaoui A (2021) Breast cancer: One-stage automated detection, segmentation, and classification of digital mammograms using UNet model based-semantic segmentation. Biomed Signal Process Control 66:102481

El Houby EM, Yassin NI (2021) Malignant and nonmalignant classification of breast lesions in mammograms using convolutional neural networks. Biomed Signal Process Control 70:102954

Saber A, Sakr M, Abo-Seida OM, Keshk A, Chen H (2021) A novel deep-learning model for automatic detection and classification of breast cancer using the transfer-learning technique. IEEE Access 9:71194–71209

Chakravarthy SS, Rajaguru H (2021) Automatic detection and classification of mammograms using improved extreme learning machine with deep learning. IRBM

Zhao J, Chen T, Cai B (2021) A computer-aided diagnostic system for mammograms based on YOLOv3. Multimed Tool Appl. https://doi.org/10.1007/s11042-021-10505-y

Kavitha T, Mathai PP, Karthikeyan C, Ashok M, Kohar R, Avanija J, Neelakandan S (2021) Deep learning based capsule neural network model for breast cancer diagnosis using mammogram images. Interdiscipl Sci Comput Life Sci 1–7.

Huang ML, Lin TY (2021) Considering breast density for the classification of benign and malignant mammograms. Biomed Signal Process Control 67:102564

Kulkarni S, Rabidas R (2022) A comparative study of different deep learning architectures for benign-malignant mass classification. In: Proceedings of the international conference on recent trends in machine learning, IoT, smart cities and applications. Springer, Singapore, pp 773–784

Oyetade IS, Ayeni JO, Ogunde AO, Oguntunde BO, Olowookere TA (2022) Hybridized deep convolutional neural network and fuzzy support vector machines for breast cancer detection. SN Comput Sci 3(1):1–4

Agarwal P, Yadav A, Mathur P (2022) Breast cancer prediction on BreakHis dataset using deep CNN and transfer learning model. In: Data engineering for smart systems. Springer, Singapore, pp 77–88

Rouhi R, Jafari M, Kasaei S, Keshavarzian P (2015) Benign and malignant breast tumors classification based on region growing and CNN segmentation. Expert Syst Appl 42(3):990–1002

Dhungel N, Carneiro G, Bradley AP (2015) Deep structured learning for mass segmentation from mammograms. In: 2015 IEEE international conference on image processing (ICIP). IEEE, pp 2950–2954

Jiao Z, Gao X, Wang Y, Li J (2016) A deep feature based framework for breast masses classification. Neurocomputing 197:221–231

Kooi T, Litjens G, Van Ginneken B, Gubern-Mérida A, Sánchez CI, Mann R, den Heeten A, Karssemeijer N (2017) Large scale deep learning for computer aided detection of mammographic lesions. Med Image Anal 35:303–312

Hepsağ PU, Özel SA, Yazıcı A (2017) Using deep learning for mammography classification. In: 2017 International conference on computer science and engineering (UBMK). IEEE, pp 418–423

Yap MH, Pons G, Marti J, Ganau S, Sentis M, Zwiggelaar R, Davison AK, Marti R (2017) Automated breast ultrasound lesions detection using convolutional neural networks. IEEE J Biomed Health Inform 22(4):1218–1226

Mendel K, Li H, Sheth D, Giger M (2019) Transfer learning from convolutional neural networks for computer-aided diagnosis: a comparison of digital breast tomosynthesis and full-field digital mammography. Acad Radiol 26(6):735–743

Sun L, Wang J, Hu Z, Xu Y, Cui Z (2019) Multi-view convolutional neural networks for mammographic image classification. IEEE Access 7:126273–126282

Samala RK, Chan HP, Hadjiiski L, Helvie MA, Richter CD, Cha KH (2018) Breast cancer diagnosis in digital breast tomosynthesis: effects of training sample size on multi-stage transfer learning using deep neural nets. IEEE Trans Med Imaging 38(3):686–696

Bressan RS, Bugatti PH, Saito PT (2019) Breast cancer diagnosis through active learning in content-based image retrieval. Neurocomputing 357:1

Dabeer S, Khan MM, Islam S (2019) Cancer diagnosis in histopathological image: CNN based approach. Inf Med Unlock. 16:100231

Li X, Radulovic M, Kanjer K, Plataniotis KN (2019) Discriminative pattern mining for breast cancer histopathology image classification via fully convolutional autoencoder. IEEE Access 7:36433–36445

Shen L, Margolies LR, Rothstein JH, Fluder E, McBride R, Sieh W (2019) Deep learning to improve breast cancer detection on screening mammography. Sci Rep 9(1):1–2

Pérez-Benito FJ, Signol F, Perez-Cortes JC, Fuster-Baggetto A, Pollan M, Pérez-Gómez B, Salas-Trejo D, Casals M, Martínez I, Lobet R (2020) A deep learning system to obtain the optimal parameters for a threshold-based breast and dense tissue segmentation. Comput Methods Prog Biomed. 195:105668

George K, Faziludeen S, Sankaran P (2020) Breast cancer detection from biopsy images using nucleus guided transfer learning and belief based fusion. Comput Biol Med 124:103954

Chang K, Beers AL, Brink L, Patel JB, Singh P, Arun NT, Hoebel KV, Gaw N, Shah M, Pisano ED, Tilkin M (2020) Multi-institutional assessment and crowdsourcing evaluation of deep learning for automated classification of breast density. J Am Coll Radiol 17(12):1653–1662

Yap MH, Goyal M, Osman F, Marti R, Denton E, Juette A, Zwiggelaar R (2020) Breast ultrasound region of interest detection and lesion localisation. Artif Intell Med 107:101880

Wang P, Song Q, Li Y, Lv S, Wang J, Li L, Zhang H (2020) Cross-task extreme learning machine for breast cancer image classification with deep convolutional features. Biomed Signal Process Control 57:101789

Singh R, Ahmed T, Kumar A, Singh AK, Pandey AK, Singh SK (2020) Imbalanced breast cancer classification using transfer learning. IEEE/ACM Trans Comput Biol Bioinf. https://doi.org/10.1109/TCBB.2020.2980831

Singh VK, Abdel-Nasser M, Akram F, Rashwan HA, Sarker MM, Pandey N, Romani S, Puig D (2020) Breast tumor segmentation in ultrasound images using contextual-information-aware deep adversarial learning framework. Expert Syst Appl 162:113870

Moon WK, Lee YW, Ke HH, Lee SH, Huang CS, Chang RF (2020) Computer-aided diagnosis of breast ultrasound images using ensemble learning from convolutional neural networks. Comput Methods Prog Biomed. 190:105361

Shu X, Zhang L, Wang Z, Lv Q, Yi Z (2020) Deep neural networks with region-based pooling structures for mammographic image classification. IEEE Trans Med Imaging 39(6):2246–2255

Li Y, Zhang L, Chen H, Cheng L (2020) Mass detection in mammograms by bilateral analysis using convolution neural network. Comput Method Prog Biomed. 195:105518

Feng Y, Zhang L, Mo J (2018) Deep manifold preserving autoencoder for classifying breast cancer histopathological images. IEEE/ACM Trans Comput Biol Bioinf 17(1):91–101

Yari Y, Nguyen TV, Nguyen HT (2020) Deep learning applied for histological diagnosis of breast cancer. IEEE Access 8:162432–162448

Wang Y, Wang N, Xu M, Yu J, Qin C, Luo X, Yang X, Wang T, Li A, Ni D (2019) Deeply-supervised networks with threshold loss for cancer detection in automated breast ultrasound. IEEE Trans Med Imaging 39(4):866–876

Zhang YD, Satapathy SC, Guttery DS, Górriz JM, Wang SH (2021) Improved breast cancer classification through combining graph convolutional network and convolutional neural network. Inf Process Manag 58(2):102439

Swiderski B, Gielata L, Olszewski P, Osowski S, Kołodziej M (2021) Deep neural system for supporting tumor recognition of mammograms using modified GAN. Expert Syst Appl 164:113968

de Lima SM, da Silva-Filho AG, Dos Santos WP (2016) Detection and classification of masses in mammographic images in a multi-kernel approach. Comput Methods Programs Biomed 134:11–29

Wang J, Yang X, Cai H, Tan W, Jin C, Li L (2016) Discrimination of breast cancer with microcalcifications on mammography by deep learning. Sci Rep 6(1):1–9

Han Z, Wei B, Zheng Y, Yin Y, Li K, Li S (2017) Breast cancer multi-classification from histopathological images with structured deep learning model. Sci Rep 7(1):1

Jiao Z, Gao X, Wang Y, Li J (2018) A parasitic metric learning net for breast mass classification based on mammography. Pattern Recogn 75:292–301

Mullooly M, Bejnordi BE, Pfeiffer RM, Fan S, Palakal M, Hada M, Vacek PM, Weaver DL, Shepherd JA, Fan B, Mahmoudzadeh AP (2019) Application of convolutional neural networks to breast biopsies to delineate tissue correlates of mammographic breast density. NPJ Breast Cancer 5(1):1–1

Yu X, Xia K, Zhang YD (2021) DisepNet for breast abnormality recognition. Comput Electr Eng 90:106961

Oyelade ON, Ezugwu AE (2020) A state-of-the-art survey on deep learning methods for detection of architectural distortion from digital mammography. IEEE Access 8:148644–148676

Shen R, Yao J, Yan K, Tian K, Jiang C, Zhou K (2020) Unsupervised domain adaptation with adversarial learning for mass detection in mammogram. Neurocomputing 393:27–37

Djebbar K, Mimi M, Berradja K, Taleb-Ahmed A (2019) Deep convolutional neural networks for detection and classification of tumors in mammograms. In: 2019 6th international conference on image and signal processing and their applications (ISPA). IEEE, pp 1–7

Cao Z, Yang Z, Zhang Y, Lin RS, Wu S, Huang L, Han M, Ma J (2019) Deep learning based mass detection in mammograms. In: GlobalSIP 2019, pp 1–5.

Mohanty F, Rup S, Dash B, Majhi B, Swamy MN (2019) Mammogram classification using contourlet features with forest optimization-based feature selection approach. Multimedia Tools and Applications 78(10):12805–12834

Agarwal R, Diaz O, Lladó X, Yap MH, Martí R (2019) Automatic mass detection in mammograms using deep convolutional neural networks. J Med Imaging 6(3):031409

Falconí LG, Pérez M, Aguilar WG (2019) Transfer learning in breast mammogram abnormalities classification with mobilenet and nasnet. In: 2019 International conference on systems, signals and image processing (IWSSIP). IEEE, pp 109–114

Tsochatzidis L, Costaridou L, Pratikakis I (2019) Deep learning for breast cancer diagnosis from mammograms—a comparative study. J Imaging 5(3):37

Zhu W, Xiang X, Tran TD, Hager GD, Xie X (2018) Adversarial deep structured nets for mass segmentation from mammograms. In: 2018 IEEE 15th international symposium on biomedical imaging (ISBI 2018). IEEE, pp 847–850

Tsochatzidis L, Zagoris K, Arikidis N, Karahaliou A, Costaridou L, Pratikakis I (2017) Computer-aided diagnosis of mammographic masses based on a supervised content-based image retrieval approach. Pattern Recogn 71:106–117

Suzuki S, Zhang X, Homma N, Ichiji K, Sugita N, Kawasumi Y, Ishibashi T, Yoshizawa M (2016) Mass detection using deep convolutional neural network for mammographic computer-aided diagnosis. In: 2016 55th annual conference of the society of instrument and control engineers of Japan (SICE). IEEE, pp 1382–1386

Mahmood T, Li J, Pei Y, Akhtar F, Imran A, Rehman KU (2020) A brief survey on breast cancer diagnostic with deep learning schemes using multi-image modalities. IEEE Access 2(8):165779–165809

Dhungel N, Carneiro G, Bradley AP (2015) Automated mass detection in mammograms using cascaded deep learning and random forests. In: 2015 international conference on digital image computing: techniques and applications (DICTA). IEEE, pp. 1–8

Dhungel N, Carneiro G, Bradley AP (2015) Tree re-weighted belief propagation using deep learning potentials for mass segmentation from mammograms. In: 2015 IEEE 12th international symposium on biomedical imaging (ISBI). IEEE, pp 760–763

Yu X, Pang W, Xu Q, Liang M (2020) Mammographic image classification with deep fusion learning. Sci Rep 10(1):1–1

Adedigba AP, Adeshinat SA, Aibinu AM (2019) Deep learning-based mammogram classification using small dataset. In 2019 15th international conference on electronics, computer and computation (ICECCO). IEEE, pp 1–6

Shen R, Yan K, Tian K, Jiang C, Zhou K (2019) Breast mass detection from the digitized X-ray mammograms based on the combination of deep active learning and self-paced learning. Futur Gener Comput Syst 101:668–679

Salem MA (2018) Mammogram-based cancer detection using deep convolutional neural networks. In: 2018 13th international conference on computer engineering and systems (ICCES). IEEE, pp 694–699

Hamidinekoo A, Denton E, Rampun A, Honnor K, Zwiggelaar R (2018) Deep learning in mammography and breast histology, an overview and future trends. Med Image Anal 47:45–67

Ismail NS, Sovuthy C (2019) Breast cancer detection based on deep learning technique. In: 2019 international UNIMAS STEM 12th engineering conference (EnCon). IEEE, pp 89–92

Gardezi SJ, Awais M, Faye I, Meriaudeau F (2017) Mammogram classification using deep learning features. In 2017 IEEE international conference on signal and image processing applications (ICSIPA). IEEE, pp. 485–488

Kausar T, MingJiang W, Ashraf MA, Kausar A (2020) SmallMitosis: small size mitotic cells detection in breast histopathology images. IEEE Access

Wang Y, Lei B, Elazab A, Tan EL, Wang W, Huang F, Gong X, Wang T (2020) Breast cancer image classification via multi-network features and dual-network orthogonal low-rank learning. IEEE Access 8:27779–27792

Benhammou Y, Achchab B, Herrera F, Tabik S (2020) BreakHis based breast cancer automatic diagnosis using deep learning: taxonomy, survey and insights. Neurocomputing 29(375):9–24

Li L, Pan X, Yang H, Liu Z, He Y, Li Z, Fan Y, Cao Z, Zhang L (2020) Multi-task deep learning for fine-grained classification and grading in breast cancer histopathological images. Multimed Tools Appl 79(21):14509–14528

Mehra R (2018) Breast cancer histology images classification: Training from scratch or transfer learning? ICT Express 4(4):247–254

Whitney HM, Li H, Ji Y, Liu P, Giger ML (2019) Comparison of breast MRI tumor classification using human-engineered radiomics, transfer learning from deep convolutional neural networks, and fusion methods. Proc IEEE 108(1):163–177

Antropova N, Huynh BQ, Giger ML (2017) A deep feature fusion methodology for breast cancer diagnosis demonstrated on three imaging modality datasets. Med Phys 44(10):5162–5171

Hu Q, Whitney HM, Giger ML (2020) A deep learning methodology for improved breast cancer diagnosis using multiparametric MRI. Sci Rep 10(1):1–1

Hai J, Tan H, Chen J, Wu M, Qiao K, Xu J, Zeng L, Gao F, Shi D, Yan B (2019) Multi-level features combined end-to-end learning for automated pathological grading of breast cancer on digital mammograms. Comput Med Imaging Graph 71:58–66

Kim EK, Kim HE, Han K, Kang BJ, Sohn YM, Woo OH, Lee CW (2018) Applying data-driven imaging biomarker in mammography for breast cancer screening: preliminary study. Sci Rep 8(1):1–8

Roty S, Wiratkapun C, Tanawongsuwan R, Phongsuphap S (2017) Analysis of microcalcification features for pathological classification of mammograms. In: 2017 10th biomedical engineering international conference (BMEiCON). IEEE, pp 1–5

Litjens G, Sánchez CI, Timofeeva N, Hermsen M, Nagtegaal I, Kovacs I, Hulsbergen-Van De Kaa C, Bult P, Van Ginneken B, Van-Der-Laak J (2016) Deep learning as a tool for increased accuracy and efficiency of histopathological diagnosis. Sci Rep 6(1):1–1

Download references

Author information

Authors and affiliations.

Department of Computer Science & Engineering, Amity School of Engineering & Technology, Amity University Rajasthan, Jaipur, India

Varsha Nemade & Sunil Pathak

Chitkara University School of Engineering and Technology, Chitkara University, Himachal Pradesh, India

Ashutosh Kumar Dubey

You can also search for this author in PubMed   Google Scholar

Corresponding author

Correspondence to Varsha Nemade .

Ethics declarations

Conflict of interest.

The authors declare no conflict of interest.

Additional information

Publisher's note.

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Nemade, V., Pathak, S. & Dubey, A.K. A Systematic Literature Review of Breast Cancer Diagnosis Using Machine Intelligence Techniques. Arch Computat Methods Eng 29 , 4401–4430 (2022). https://doi.org/10.1007/s11831-022-09738-3

Download citation

Received : 09 November 2021

Accepted : 23 March 2022

Published : 11 April 2022

Issue Date : October 2022

DOI : https://doi.org/10.1007/s11831-022-09738-3

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

  • Find a journal
  • Publish with us
  • Track your research

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

The PMC website is updating on October 15, 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • v.64; 2022 Aug

Global guidelines for breast cancer screening: A systematic review ☆

a Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China

Mingyang Chen

b Center for Global Health, School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China

Youlin Qiao

Fanghui zhao.

Breast cancer screening guidelines could provide valuable tools for clinical decision making by reviewing the available evidence and providing recommendations. Little information is known about how many countries have issued breast cancer screening guidelines and the differences among existing guidelines. We systematically reviewed current guidelines and summarized corresponding recommendations, to provide references for good clinical practice in different countries.

Systematic searches of MEDLINE, EMBASE, Web of Science, and Scopus from inception to March 27th, 2021 were conducted and supplemented by reviewing the guideline development organizations. The quality of screening guidelines was assessed from six domains of the Appraisal of Guidelines for Research and Evaluation Ⅱ (AGREE Ⅱ) instrument by two appraisers. The basic information and recommendations of the issued guidelines were extracted and summarized.

A total of 23 guidelines issued between 2010 and 2021 in 11 countries or regions were identified for further review. The content and quality varied across the guidelines. The average AGREE Ⅱ scores of the guidelines ranged from 33.3% to 87.5%. The highest domain score was "clarity of presentation" while the domain with the lowest score was "applicability". For average-risk women, most of the guidelines recommended mammographic screening for those aged 40–74 years, specifically, those aged 50–69 years were regarded as the optimal age group for screening. Nine of 23 guidelines recommended against an upper age limit for breast cancer screening. Mammography (MAM) was recommended as the primary screening modality for average-risk women by all included guidelines. Most guidelines suggested annual or biennial mammographic screening. Risk factors of breast cancer identified in the guidelines mainly fell within five categories which could be broadly summarized as the personal history of pre-cancerous lesions and/or breast cancer; the family history of breast cancer; the known genetic predisposition of breast cancer; the history of mantle or chest radiation therapy; and dense breasts. For women at higher risk, there was a consensus among most guidelines that annual MAM or annual magnetic resonance imaging (MRI) should be given, and the screening should begin earlier than the average-risk group.

Conclusions

The majority of 23 included international guidelines were issued by developed countries which contained roughly the same but not identical recommendations on breast cancer screening age, methods, and intervals. Most guidelines recommended annual or biennial mammographic screening between 40 and 74 years for average-risk populations and annual MAM or annual MRI starting from a younger age for high-risk populations. Current guidelines varied in quality and increased efforts are needed to improve the methodological quality of guidance documents. Due to lacking clinical practice guidelines tailored to different economic levels, low- and middle-income countries (LMICs) should apply and implement the evidence-based guidelines with higher AGREE Ⅱ scores considering local adaption.

  • • This systematic review comprehensively maps the recommendations of the latest international breast screening guidelines, providing valuable tools for clinical decision making in different settings.
  • • Most guidelines recommend annual or biennial mammographic screening between 40 and 74 years for the average-risk populations and annual MAM or annual MRI starting from a younger age for the high-risk populations. However, there are indeed discrepancies in screening age, methods, and intervals among countries.
  • • High-quality evidence and rigorous methodology are the keys to guidance development, but current guidelines vary in methodological quality.

1. Introduction

In 2021, breast cancer has overtaken lung cancer to be the world's most commonly diagnosed cancer, accounting for the severe burden globally, especially among women [ 1 ]. Screening for breast cancer is an effective measure to detect early-stage disease and improve the survival rate of cancer patients [ [2] , [3] ]. Population-based breast cancer screening programs have been implemented in many developed countries over the last decades, which contributed to reducing the mortality and the advanced cancer rate [ [4] , [5] , [6] ].

Screening guidelines could provide valuable tools for clinical decision making by reviewing the available evidence and providing recommendations. To date, several breast cancer screening guidelines have been issued in many developed countries [ [7] , [8] , [9] ]. However, the recommendations about screening age, methods, and intervals varied from different guidelines due to different institutions, based evidence, and development processes. This may confuse the clinical practice when they are applied to other countries. To our knowledge, it is currently unknown how many countries have issued breast cancer screening guidelines and the differences among these issued guidelines. Additionally, previous systematic reviews of international breast cancer screening guidelines were limited by publication date and screening population and did not systematically review screening recommendations for the population with different breast cancer risks [ [10] , [11] , [12] ].

Accordingly, our study reviewed existing breast cancer screening guidelines and summarized corresponding recommendations, in order to provide references for good clinical practice in different countries.

2. Material and methods

2.1. data sources and searches.

A search strategy was designed for MEDLINE, EMBASE, Web of Science, and Scopus from inception to March 27th, 2021 using variations on the search terms "breast cancer", "screening" and "guidelines/recommendations" ( Appendix A ). We also sought the additional guidelines by searching guideline development organizations, such as Guideline International Network (GIN), World Health Organization (WHO), Cancer Australia, Ministry of Health (MOH) Malaysia, and China Guideline Clearinghouse (CGC). Moreover, we meticulously examined the references of documents obtained above to further access potentially eligible articles.

2.2. Study selection and data extraction

The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) flow diagram is presented in Fig. 1 . Two reviewers (MYC and WHR) independently reviewed the titles and abstracts of the included guidelines. Any discrepancies were resolved by discussion. Finally, both reviewers determined the included guidelines based on the full text. We included guidelines following inclusion criteria: (1) originally published guidelines, consensus, or position papers related to breast cancer screening; (2) the latest versions of the updated guidelines; (3) English or Chinese guidelines; and (4) full text was available. We excluded guidelines if they were: summaries or interpreted versions of guidelines.

Fig. 1

Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) flow diagram.

Two independent reviewers (MYC and WHR) extracted information using a predesigned template. The information extracted included: (1) basic information (countries or regions, publication years, publication organizations, names of guidelines, number of updated versions, and publication years of old versions); (2) screening recommendations for the population at average risk and higher risk (screening age, screening methods, screening intervals, level of evidence, and strength of recommendation).

2.3. Quality assessment

The methodological quality of guidelines was evaluated using the Appraisal of Guidelines for Research and Evaluation Ⅱ (AGREE Ⅱ) instrument. This is a standardized tool for evaluating the methodological framework of guideline development which consists of 23 main items in six domains (scope and purpose, stakeholder involvement, rigour of development, clarity of presentation, applicability, and editorial independence) and two global rating items [ 13 ]. Each item is rated on a seven-point Likert-type scale from one (strongly disagree) to seven (strongly agree) according to the criteria and considerations articulated in the User's Manual. Scores are assigned depending on the completeness and quality of reporting. Scores increase as more criteria are met and considerations are addressed. Domain scores are calculated by summing up all the scores of the individual items in that domain and by scaling the total as a percentage of the maximum possible score for that domain. Two reviewers (MYC and WHR) independently scored each guideline. Evaluation results were compared and discrepancies of more than two points per item were discussed to reach a consensus. According to prior studies, the quality of guidelines was classified as high if the total score was 60% or higher and low if the score was less than 60% [ 14 , 15 ].

A total of 7417 citations were included during the preliminary literature search process, but most were excluded after deleting duplicates and applying the inclusion and exclusion criteria. Of these, 23 guidelines were identified for further review ( Fig. 1 ).

3.1. Guideline characteristics

Table 1 displays the general characteristics of 23 included guidelines that were published between 2010 and 2021 [ [7] , [8] , [9] , [16] , [17] , [18] , [19] , [20] , [21] , [22] , [23] , [24] , [25] , [26] , [27] , [28] , [29] , [30] , [31] , [32] , [33] , [34] , [35] ]. The majority of guidelines (17 of 23) were drawn from developed countries or regions. Guidelines from the United States accounted for the largest proportion, reaching 39.1%. One was developed by WHO, and four in Europe ( Fig. 2 ). 12 of 23 guidelines have been updated.

Characteristics of 23 included guidelines on screening for breast cancer.

Countries/RegionsPublication yearsPublication organizationsNames of guidelinesNumber of updated versionsPublication years of old versions
Global [ ]2014WHOWHO position paper on mammography screeningNoneNone
The United States [ ]2019ACPScreening for Breast Cancer in Average-Risk Women: A Guidance Statement From the American College of Physicians12007
The United States [ ]2019NCCNBreast Cancer Screening and Diagnosis, Version 1.201991998 2003 2006 2010 2013 2015 2016 2017 2018
The United States [ ]2017ACRACR Appropriateness Criteria® Breast Cancer Screening21998 2013
The United States [ ]2018ACRBreast Cancer Screening in Women at Higher-Than-Average Risk: Recommendations From the ACRNoneNone
The United States [ ]2017ACRBreast Cancer Screening for Average-Risk Women: Recommendations From the ACR Commission on Breast ImagingNoneNone
The United States [ ]2010ACR and SBIBreast Cancer Screening With Imaging: Recommendations From the Society of Breast Imaging and the ACR on the Use of Mammography, Breast MRI, Breast Ultrasound, and Other Technologies for the Detection of Clinically Occult Breast CancerNoneNone
The United States [ ]2016USPSTFScreening for Breast Cancer: U.S. Preventive Services Task Force Recommendation Statement31996 2002 2009
The United States [ ]2015ACSBreast Cancer Screening for Women at Average Risk 2015 Guideline Update From the American Cancer Society31992 1997 2003
The United States [ ]2019ACOGBreast Cancer Risk Assessment and Screening in Average-Risk Women32003 2011
2017
Europe [ ]2020ECIBCBreast Cancer Screening and Diagnosis: A Synopsis of the European Breast GuidelinesNoneNone
Europe [ ]2019ESMOEarly breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up12015
Europe [ ]2010EUSOMAMagnetic resonance imaging of the breast: Recommendations from the EUSOMA working groupNoneNone
Canada [ ]2018CTFPHCRecommendations on screening for breast cancer in women aged 40–74 years who are not at increased risk for breast cancer31994 2001 2011
Germany [ ]2018AWMF, DKG, and DKHThe Screening, Diagnosis, Treatment, and Follow-Up of Breast Cancer12012
Australia [ ]2015Cancer AustraliaEarly detection of breast cancer22009 2004
Singapore [ ]2010MOHCancer screeningNoneNone
Malaysia [ ]2019MOHManagement of Breast Cancer (3rd Edition)22002 2010
Japan [ ]2016NCC JapanThe Japanese Guidelines for Breast Cancer Screening22013 2015
China [ ]2021NCC ChinaChina Guideline for the Screening and Early Detection of Female Breast Cancer (2021, Beijing)NoneNone
Hong Kong, China [ ]2018CEWGRecommendations on prevention and screening for breast cancer in Hong KongNoneNone
Brazil [ ]2018MOHGuidelines for early detection of breast cancer in Brazil. II – New national recommendations, main evidence, and controversiesNoneNone
Brazil [ ]2017CBR, SBM, and FEBRASGOBreast Cancer Screening: Updated Recommendations of the Brazilian College of Radiology and Diagnostic Imaging, Brazilian Breast Disease Society, and Brazilian Federation of Gynecological and Obstetrical AssociationsNoneNone

Abbreviations: ACOG: American College of Obstetricians and Gynecologists; ACP: American College of Physicians; ACR: American College of Radiology; ACS: American Cancer Society; AWMF: German Association of Scientific Medical Societies; CBR: Brazilian College of Radiology and Diagnostic Imaging; CEWG: Cancer Expert Working Group; CTFPHC: Canadian Task Force on Preventive Health Care; DKG: German Cancer Society; DKH: German Cancer Aid; ECIBC: European Commission Initiative on Breast Cancer; ESMO: European Society for Medical Oncology; EUSOMA: European Society of Breast Cancer Specialists; FEBRASGO: Brazilian Federation of Gynecological and Obstetrical Associations; MOH: Ministry of Health; NCC: National Cancer Centre; NCCN: National Comprehensive Cancer Network; SBI: Society of Breast Imaging; SBM: Brazilian Society for Breast Disease; USPSTF: U.S. Preventive Services Task Force; WHO: World Health Organization.

Fig. 2

Geographical distribution of the included breast cancer screening guidelines.

3.2. Quality assessment

The included 23 guidelines were appraised using AGREE II Criteria ( Fig. 3 ). The average AGREE II scores varied from 33.3% to 87.5%. 12 guidelines were scored over 60.0% [ [7] , [8] , [16] , [17] , [23] , [24] , [25] , [27] , [30] , [31] , [32] , [34] ]. Among these, the guideline issued by Canadian Task Force on Preventive Health Care (CTFPHC) [ 27 ] was scored the highest (87.5%), followed by European Commission Initiative on Breast Cancer (ECIBC) [ 25 ], American Cancer Society (ACS) [ 23 ], United States Preventive Services Taskforce (USPSTF) [ 7 ], and WHO [ 16 ]. The highest domain score was "clarity of presentation" (domain 4), with an average score of 81.9%, followed by "scope and purpose" (domain 1). The domain with the lowest score was "applicability" (domain 5) with an average score of 21.3%, followed by "stakeholder involvement" (domain 2).

Fig. 3

Quality of the included guidelines for the six domains of the AGREE Ⅱ instrument.

( Abbreviations: ACOG: American College of Obstetricians and Gynecologists; ACP: American College of Physicians; ACR: American College of Radiology; ACS: American Cancer Society; AWMF: German Association of Scientific Medical Societies; CBR: Brazilian College of Radiology and Diagnostic Imaging; CEWG: Cancer Expert Working Group; CTFPHC: Canadian Task Force on Preventive Health Care; ECIBC: European Commission Initiative on Breast Cancer; ESMO: European Society for Medical Oncology; EUSOMA: European Society of Breast Cancer Specialists; MOH: Ministry of Health; NCC: National Cancer Centre; NCCN: National Comprehensive Cancer Network; SBI: Society of Breast Imaging; USPSTF: U.S. Preventive Services Task Force; WHO: World Health Organization).

3.3. Strength of recommendations and quality of evidence

17 of 23 guidelines reported eight applied grading systems. Grading of Recommendations, Assessment, Development and Evaluations (GRADE) was the common system that was applied in six guidelines [ 16 , 23 , 25 , 27 , 32 , 34 ]. Four guidelines used the self-designated grading system [ 8 , 9 , 18 , 30 ]. The details about the strength of recommendations and the quality of evidence varied in different grading systems. The information of evidence and recommendation about the included guidelines is shown in Table 2 .

Grading systems used in the included guidelines.

Grading systemsGuideline organizationsLevel of evidenceStrength of recommendations
GRADEWHO, 2014 [ ]; ACS, 2015 [ ]; ECIBC, 2020 [ ]; CTFPHC, 2018 [ ]; MOH of Brazil, 2018 [ ]; NCC China, 2021 [ ]High; Moderate; Low; Very lowStrong; Qualified/Conditional; Weak
GRADE + RAMACR, 2017 [ ]Strong; Moderate; LimitedUsually appropriate; May be appropriate; Usually not appropriate
USPSTFUSPSTF, 2010 [ ]; ACOG, 2019 [ ]Ⅰ; Ⅱ-1; Ⅱ-2; Ⅱ-3; ⅢA; B; C; D; I (insufficient)
USPSTF + GRADEMOH of Malaysia, 2019 [ ]Ⅰ; Ⅱ-1; Ⅱ-2; Ⅱ-3; ⅢStrong;
Conditional
OCEBMEUSOMA, 2010 [ ]; AWMF, DKG, and DKH, 2018 [ ]1 a/1 b/1 c; 2 a/2 b; 3 a/3 b; 4; 5A; B; C; D
OCEBM + GRADECBR, SBM, and FEBRASGO, 2018 [ ]NoneA; B; C; D
NCCNNCCN, 2019 [ ]1; 2 A; 2 B; 3A; B; C; D
Adapted from the Infectious Disease Society of America-United States Public Health Service Grading SystemESMO, 2019 [ ]Ⅰ; Ⅱ; Ⅲ; Ⅳ; ⅤA; B; C; D; E
JRGCSGNCC Japan, 2016 [ ]NoneA; B; C; D; I (insufficient)
MOH, SingaporeMOH of Singapore, 2010 [ ]1++; 1+; 1-; 2++; 2+; 2-;3; 4A; B; C; D; GPP

Abbreviations: ACOG: American College of Obstetricians and Gynecologists; ACS: American Cancer Society; ACR: American College of Radiology; AWMF: German Association of Scientific Medical Societies; CBR: Brazilian College of Radiology and Diagnostic Imaging; CTFPHC: Canadian Task Force on Preventive Health Care; DKG: German Cancer Society; DKH: German Cancer Aid; ECIBC: European Commission Initiative on Breast Cancer; ESMO: European Society for Medical Oncology; EUSOMA: European Society of Breast Cancer Specialists; FEBRASGO: Brazilian Federation of Gynecological and Obstetrical Associations; GPP: Good Practice Points; GRADE: Grading of Recommendations, Assessment, Development and Evaluations; JRGCSG: Japanese Research Group for the Development of Cancer Screening Guidelines; MOH: Ministry of Health; NCC: National Cancer Centre; NCCN: National Comprehensive Cancer Network; OCEBM: Oxford Centre for Evidence-based Medicine; RAM: RAND/UCLA Appropriateness Method; SBM: Brazilian Society for Breast Disease; USPSTF: U.S. Preventive Services Task Force; WHO: World Health Organization.

3.4. The screening recommendations for women at average risk

The detailed information of recommendations for average-risk women is shown in Table 3 , which summarized screening age, screening methods, screening intervals, and other recommended screening methods ( Fig. 4 ).

The screening recommendations in average-risk women in eligible guidelines.

GuidelinesAge range for screeningAge to end screeningScreening methodsScreening intervalsRecommendations for other screening methods
WHO, 2014 [ ]40–49 years; 70-74 yearsNRMAM NRNR
50–69 years CBE seems to be a promising approach in limited resource settings with weak health systems
ACP, 2019 [ ]40–49 years≥ 75 years or in women with a life expectancy of 10 years or lessNR NRNot recommend CBE
50–74 yearsMAMBiennial
NCCN, 2019 [ ]25–39 years Clinical encounter Every 1–3 years
≥ 40 yearsClinical encounter Annual
MAM Annual
ACR, 2017 [ ]≥ 40 yearsNRMAM or DBTAnnualFor women with dense breasts, US may be considered, but the increased cancer detection and the increased risk of a false-positive examination should be weighed
ACR (Average-risk), 2017 [ ]≥ 40 yearsThe age to stop screening should be based on each woman's health status rather than an age-based determinationMAMAnnualNo sufficient data to support the use of breast MRI and MBI as a screening tool for average-risk women
ACR and SBI, 2010 [ ]≥ 40 years MAMAnnualNR
USPSTF, 2016 [ ]40–49 years75 years MAM Biennial
50–74 yearsMAM Biennial
ACS, 2015 [ ]40–44 yearsScreening should continue as long as a woman is in good health and is expected to live at least 10 more yearsMAM Annual Not recommend CBE
45–54 yearsMAM Annual
≥ 55 yearsMAM Annual or biennial
ACOG, 2019 [ ]25–39 years CBE Every 1–3 years
≥ 40 yearsMAM Annual or biennial
Biennial (after age 55)
CBE Annual
ECIBC, 2020 [ ]45–49 yearsNRMAM
50–69 yearsMAM
70–74 yearsMAM
ESMO, 2019 [ ]40–49 years; 70–74yearsNRMAM [B]NRNR
50–69 yearsMAM [A]Annual or biennial [A]
CTFPHC, 2018 [ ]50–74 yearsNRMAM Every 2–3 years
AWMF, DKG, and DKH, 2020 [ ]50–69 years≥ 70 years: taking into consideration their individual risk profile and health status, as well as a life expectancy of more than 10 yearsMAMBiennialInsufficient evidence about other imaging examination (tomosynthesis, US, MRI, or other techniques) contributes to a reduction in breast cancer mortality, neither as a supplemental examination nor a substitute for MAM
Cancer Australia, 2015 [ ]40–49 years≥ 75 years: be eligible to receive free MAM, but do not receive an invitation to attendMAM (discuss, by SDM)NRNo evidence to recommend for or against CBE
50–74 yearsMAMBiennial
MOH of Singapore, 2010 [ ]40–49 years≥70 years: be individualized by considering the potential benefits and risks of mammography in the context of current health status and estimated life expectancyMAM Annual US and CBE are not routinely required
50–69 yearsMAM Biennial
MOH of Malaysia, 2019 [ ]50–74 yearsNRMAMBiennialNR
NCC Japan, 2016 [ ]40–64 yearsNRMAM with CBENRCBE and US are not recommended for population-based screening
40–74 yearsMAM without CBE
NCC China, 2021 [ ]≥ 45 yearsNR Annual or biennial
MOH of Brazil, 2018 [ ]50–69 years MAM Biennial
CBR, SBM, and FEBRASGO, 2017 [ ]40–74 years≥ 75 years MAM (preferably digital MAM) Annual

Abbreviations: ABUS: Automated Breast Ultrasonography; ACOG: American College of Obstetricians and Gynecologists; ACP: American College of Physicians; ACR: American College of Radiology; ACS: American Cancer Society; AWMF: German Association of Scientific Medical Societies; BSE: Breast Self-Examination; CBE: Clinical Breast Examination; CBR: The Brazilian College of Radiology and Diagnostic Imaging; CTFPHC: Canadian Task Force on Preventive Health Care; DKG: German Cancer Society; DKH: German Cancer Aid; DBT: Digital Breast Tomosynthesis; ECIBC: European Commission Initiative on Breast Cancer; ESMO: European Society for Medical Oncology; FEBRASGO: Brazilian Federation of Gynecological and Obstetrical Associations; HHUS: Hand-Held Ultrasound; MAM: Mammography; MBI: Molecular Breast Imaging; MOH: Ministry of Health; MRI: Magnetic Resonance Imaging; NCC: National Cancer Centre; NCCN: National Comprehensive Cancer Network; NR: No Recommendation; SBM: The Brazilian Society for Breast Disease; SDM: Shared Decision Making; US: Ultrasound; USPSTF: U.S. Preventive Services Task Force; WHO: World Health Organization.

Fig. 4

The main screening recommendations in average-risk women in the eligible guidelines.

( Abbreviations: CBE: Clinical Breast Examination; MAM: Mammography; US: Ultrasound)

3.4.1. Screening age

The majority of guidelines recommended mammographic screening for average-risk individuals aged 40–74 years [ [7] , [8] , [9] , 16 , 17 , 29 , 35 ], and recommended women aged 50–69 years as the optimal age group for screening with strong recommendation [ 8 , 16 , 25 , 28 , 30 , 34 ]. National Comprehensive Cancer Network (NCCN) [ 18 ] and American College of Obstetricians and Gynecologists (ACOG) [ 24 ] suggested starting screening at age 25 by clinical encounter or clinical breast examination (CBE).

Nine of 23 guidelines did not recommend an upper age limit for breast cancer screening [ 8 , 9 , 16 , 18 , 20 , 25 , 27 , 31 , 32 ]. Some guidelines, including American College of Radiology (ACR) [ 21 ], ACR and Society of Breast Imaging (SBI) [ 22 ], and ACS [ 23 ] suggested that the age to end screening should be determined based on the women's health status, for example, stopping screening for women with life expectancy lower than 5–7 years or 10 years. Other guidelines, like USPSTF [ 7 ], American College of Physicians (ACP) [ 17 ], and Brazilian College of Radiology and Diagnostic Imaging (CBR)/Brazilian Society for Breast Disease (SBM)/Brazilian Federation of Gynecological and Obstetrical Associations (FEBRASGO) [ 35 ] did not recommend breast cancer screening for women aged over 75 years unless their life expectancy were higher than 7 years or 10 years. German Association German Cancer Society of Scientific Medical Societies (AWMF)/German Cancer Society (DKG)/German Cancer Aid (DKH) [ 28 ] and MOH of Singapore [ 30 ] recommended stopping screening at age 70.

3.4.2. Screening methods and intervals

Mammography (MAM) was recommended as the primary screening modality for average-risk women by all included guidelines [ [7] , [8] , [9] , [16] , [17] , [18] , [19] , [21] , [22] , [23] , [24] , [25] , [26] , [27] , [28] , [29] , [30] , [31] , [32] , [34] , [35] ]. Most guidelines suggested annual or biennial mammographic screening [ 7 , 16 , 17 , 29 , 31 ]. Three guidelines recommended screening every 1–2 years [ [8] , [23] , [32] ]. Some guidelines agreed that screening intervals should be determined based on age [ 18 , 24 ]. ACS [ 23 ] recommended screening with MAM annually for women aged 40–54 years and every 1–2 years for women aged 55 years or older. ECIBC [ 25 ] recommended screening every 2–3 years for women aged 40–49 years and for women aged 70–74 years. For the priority screening groups (women aged 50–69 years), annual screening was not recommended, and biennial screening is better than triennial screening.

The recommendations of each guideline on CBE and ultrasound (US) were different in detail. NCCN [ 18 ] and ACOG [ 24 ] suggested that CBE should be given every 1–3 years for women aged 25–39 years and annually for women older than 40 years, but ACS [ 23 ] and CTFPHC [ 27 ] did not recommend CBE as a primary screening method. Among the included screening guidelines, only National Cancer Centre (NCC) of China [ 32 ] recommended screening every 1–2 years for women older than 45 years using US alone.

All guidelines did not recommend using breast self-examination (BSE), magnetic resonance imaging (MRI), and computed tomography (CT) to screen for average-risk women because of lacking evidence of benefit.

3.5. The screening recommendations for women at higher risk

Risk factors of breast cancer identified in the guidelines mainly fell within five categories which could be broadly summarized as the personal history of pre-cancerous lesions and/or breast cancer; the family history of breast cancer; the known genetic predisposition of breast cancer; the history of mantle or chest radiation therapy; and dense breasts. For women at higher risk, there was a consensus among most guidelines that annual MAM screening or annual MRI screening should be given and the starting age should be earlier than the average-risk group ( Table 4 ; Fig. 5 ) .

The screening recommendations in high-risk women in eligible guidelines.

Risk factorsGuidelinesScreening ageScreening methods and intervals
]
ACR (High-risk), 2017 [ ]From the time of diagnosisMRI: annual
]
MOH of Singapore, 2010 [ ]NRMAM: annual [Grade D]
MOH of Malaysia, 2019 [ ]40–59 years, 30–39 years (may be considered)MAM: annual
≥60 yearsMAM: biennial
NCC China, 2021 [ ]NRMAM and US: annual
CEWG, 2018 [ ]From 35 yearsMAM: annual
CBR, SBM, and FEBRASGO, 2017 [ ]
Family history of breast cancer ]
ESMO, 2019 [ ]NRMAM and MRI: annual (concomitant or alternating) [Ⅲ, A]
NCC China, 2020 [ ]NRMAM and US: annual
CEWG, 2018 [ ]Begin at age 35 or 10 years prior to the age at diagnosis of the youngest-affected relative (for those with family history), whichever is earlier, but not earlier than age 30.MAM: annual
Known genetic predisposition of breast cancer ]
ACR (High-risk), 2017 [ ]30 yearsDM+/DBT: annual
25–30 yearsMRI: annual
ACR and SBI, 2010 [ ]Start by age 30 but not before age 25MAM: annual
EUSOMA, 2010 [ ]Start from 30 years;
Before 30 years [discuss, mutation carrier of BRCA1 or BRCA2 (start from 25 to 29) and TP53 (start from 20)]
MRI: annual
MOH of Singapore, 2010 [ ]Start at age 25–30 years for BRCA mutation carriers and their untested first-degree relatives, or as early as 5–10 years before the age of onset of breast cancer in the youngest family member in those with family history of breast cancer but no proven mutation
MOH of Malaysia, 2019 [ ]30–49 yearsMRI: annual
40–69 yearsMAM: annual
≥70 yearsMAM: biennial
NCC China, 2020 [ ]NRMRI: annual
CBR, SBM, and FEBRASGO, 2017 [ ]From 30 yearsMAM annual [category B recommendation]
From 25 yearsMRI annual [category A recommendation]
History of mantle or chest radiation therapyNCCN, 2019 [ ]Start from 10 years after radiation exposureBreast awareness and clinical encounter: every 6–12 months
Start from 10 years after radiation exposure but not less than age 30DM: annual, with consideration of tomosynthesis
Start from 10 years after radiation exposure but not less than age 25MRI: annual
Start from 10 years after radiation exposure for women younger than 25 years who have received prior thoracic irradiationBreast awareness, counseling on risk and an annual clinical encounter
ACR, 2017 [ ]Start from age 25 or 8 years after radiation therapy, whichever is laterMAM
ACR (High-risk), 2017 [ ]Start from age 25 or 8 years after radiation therapy, whichever is laterDM+/DBT: annual
NCC China, 2020 [ ]NRMRI: annual
]
CBR, SBM, and FEBRASGO, 2017 [ ]Start from the 8th year after radiotherapy onward, but not begin before age 30MAM: annual [category C recommendation]
Start from the 8th year after radiotherapy onward, but not begin before age 25MRI: annual [category C recommendation]
Dense breastsACR, 2017 [ ]NRConsider: US
ACR (High-risk), 2017 [ ]NR
ACR and SBI, 2010 [ ]NRConsider: US as an adjunct to MAM
China NCC, 2020 [ ]NRMAM and US: annual
CBR, SBM, and FEBRASGO, 2017 [ ]NRConsider: US as an adjunct to MAM

Abbreviations: ACR: American College of Radiology; BRCA: Breast cancer gene; BSE: Breast Self-Examination; CBE: Clinical Breast Examination; CBR: Brazilian College of Radiology and Diagnostic Imaging; CEWG: Cancer Expert Working Group; DBT: Digital Breast Tomosynthesis; DM: Digital Mammography; ESMO: European Society for Medical Oncology; EUSOMA: European Society of Breast Cancer Specialists; FEBRASGO: Brazilian Federation of Gynecological and Obstetrical Associations; MAM: mammography; MOH: Ministry of Health; MRI: Magnetic Resonance Imaging; NCC: National Cancer Centre; NCCN: National Comprehensive Cancer Network; NR: No Recommendation; SBI: Society of Breast Imaging; SBM: Brazilian Society for Breast Disease; US: Ultrasound.

Fig. 5

The main screening recommendations in high-risk women in the eligible guidelines.

( Abbreviations: BSE: Breast Self Examination; CBE: Clinical Breast Examination; MAM: Mammography; MRI: Magnetic Resonance Imaging; NR: No Recommendation; US: Ultrasound)

3.5.1. Women with the personal history of pre-cancerous lesions and/or breast cancer

For women with biopsy-proven Lobular Carcinoma in Situ (LCIS), Atypical Ductal Hyperplasia (ADH), Ductal Carcinoma in Situ (DCIS), or invasive breast cancer or ovarian cancer, annual MAM or annual MRI was mainly recommended after diagnosis onward [ 18 , 20 , 22 , 31 , 33 , 35 ] . Especially for patients with unilateral invasive breast cancer, close monitoring of the contralateral breast was recommended. NCCN [ 18 ] also recommended breast awareness and clinical encounter every 6–12 months for this group of women. NCC of China [ 32 ] recommended MAM and US as screening methods for women at higher risk of breast cancer.

3.5.2. Women with the family history of breast cancer

For women with a family history suspicious of the inherited predisposition of breast cancer, two guidelines recommended that an annual MAM or annual MRI began 10 years before the age of diagnosis of the youngest-affected relative but not before the age of 30 [ 18 , 33 ]. NCCN [ 18 ] also recommended regular clinical visits every 6–12 months once the women were identified as begin at increased risk of breast cancer.

3.5.3. Women with the known genetic predisposition of breast cancer

Women with breast cancer susceptibility gene 1 (BRCA1) or breast cancer susceptibility gene 2 (BRCA2) mutations, or untested but have first-degree relatives (mothers, sisters, or daughters) who are proven to have BRCA mutations, have a higher risk for breast cancer. Two guidelines recommended that women with gene mutations should start to undertake annual MAM or annual MRI at 25–30 years [ 22 , 30 ]. European Society of Breast Cancer Specialists (EUSOMA) [ 26 ] recommended that annual MRI screening was performed for women carrying BRAC at 25–29 years, and those carrying TP53 at 20 years. MOH of Malaysia [ 31 ] provided age-specific recommendations for women carrying gene mutations, specifically, annual MRI for 30–49 years, annual MAM for 40–69 years, and biennial MAM for 70 years and above. For other recommended screening methods, ACR [ 19 ] recommended MRI as an adjunct to MAM or DBT and recommended US when the patient cannot tolerate MRI. MOH of Singapore [ 30 ] also recommended monthly BSE and 6 monthly CBE.

3.5.4. Women with the history of mantle or chest radiation therapy

For women with a history of mantle or chest radiation therapy that occurred before the age of 30 years or had a cumulative dose of 10 Gy radiation, most guidelines recommended starting regular screening 8 or 10 years after radiation therapy [ [18] , [19] , [20] , 35 ]. Recommended screening strategies included annual MAM (not before age 30), annual MRI (not before age 25), or annual digital mammography (DM) (with or without digital breast tomosynthesis (DBT)). NCCN [ 18 ] also recommended increasing breast awareness or clinical encounters every 6–12 months.

3.5.5. Women with dense breasts

For women with dense breasts, ACR [ 20 ] recommended MRI should be performed annually. NCC of China [ 32 ] recommended screening with MAM and US annually. US (as adjunctive screening tools) was recommended for high-risk women who may be suitable for MRI but can not be accepted for any reason [ 20 ]. Two guidelines [ [22] , [35] ] also recommended US as an adjunctive examination to MAM in asymptomatic women with dense breasts.

4. Discussion

To the best of our knowledge, this study is the largest and most comprehensive systematic review, which identified and compared the latest international breast screening guidelines and recommendations. A total of 23 guidelines issued between 2010 and 2021 in 11 countries or regions were included in this study. The content and quality varied between the guidelines. The average AGREE Ⅱ scores ranged from 33.3% to 87.5%, which is consistent with that reported by Li J et al. [ 12 ]. We found discrepancies between guidelines concerning screening age, methods, and intervals. In general, the majority of guidelines agreed upon annual or biennial MAM for average-risk women aged 40 to 74. Annual MAM or annual MRI should be given and start earlier for women at high risk for breast cancer.

Our study showed that many low- and middle-income countries (LMICs) lacked published clinical practice guidelines for breast cancer screening. Most included guidelines in our study were issued by developed countries, mainly in the United States (9/23) and Europe (4/23). One possible explanation is that high-income countries have accumulated more high-quality evidence for developing guidelines by implementing breast cancer screening programs and related research for a long time [ [4] , [5] , [6] ]. However, although LMICs have a severe breast cancer burden, few tailored guidelines have been issued due to lacking sufficient national evidence about breast cancer screening and the front-line impact of sparse resources to develop guidelines in these areas [ 36 , 37 ]. Additionally, some LMICs guidelines might be published in local languages and were not picked up in our search. We also found that some guidelines issued by LMICs are often based on evidence from high-income countries. The extent to which these guidelines can be applied to the clinical practice of routine screening in LMICs is unknown.

High-quality guidelines are vital to facilitate clinical decision making and to improve health outcomes and health service efficiency. Our findings showed nearly half of the included guidelines were rated as high quality. Most of the guidelines provided a clear description of "scope and purpose" as screening for populations with different breast cancer risks, and screening recommendations were described clearly. For these reasons, the domains "scope and purpose" and "clarity of presentation" received high scores. In contrast, the majority of the guidelines received low scores in the domains of "rigour of development" and "applicability". According to prior studies [ [38] , [39] ], the domain "rigour of development" was the most relevant to the overall quality of the guideline. The main reason was that this domain reflects the evidence collection and synthesis process, as well as the formation and follow-up update of recommendations, which can provide enough information to evaluate whether the guidelines followed the best methodology and developed evidence-based recommendations. Meanwhile, the development process of guidelines is also one of the key reasons causing the variations between the recommendations from different guidance documents. In our study, 17 of 23 guidelines reported using eight different grading systems to evaluate the quality of evidence and strength of recommendations, which somewhat impeded the implementation of the guidelines and caused confusion in clinical practice. The most important purpose of guidelines is to promote their application to real-world medicine practice. Therefore, guideline developers should clearly describe the promotion conditions and hindrance factors in the implementation of recommendations and their improvement strategies, as well as consider the likely resource implications involved. At the same time, the quality of the "applicability" domain also plays a critical role in whether they can be extended to LMICs that might lack indigenous guidelines. Our study showed that the scores of different guidelines varied greatly in the domain "applicability". For example, the guideline issued by MOH of Malaysia [ 31 ] contained a separate section called "implementing the guidelines", which described the types of facilitators and barriers in detail, as well as put forward suggestions to ensure the implementation of the guideline. In contrast, the "ACR Appropriateness Criteria® Breast Cancer Screening" [ 19 ] did not mention facilitators and barriers to its application. Based on the above considerations, we considered the guideline developed by MOH of Malaysia with high "applicability" rather than ACR.

The majority of guidelines recommended mammographic screening for average-risk women aged 40–74 years. 50–69 years were regarded as the optimal age group for screening due to the steep increase of breast cancer beginning around age 50. In 2019, almost 82% of breast cancer was diagnosed among women aged ≥ 50 years in the United States [ 40 ]. Most randomized controlled trials (RCTs) from developed countries also showed that mammographic screening between 50 and 69 years had the greatest benefit in reducing mortality [ [41] , [42] ]. However, due to the disease burden of breast cancer and the allocation of public health resources vary in different countries, a one-size-fits-all approach to screening is considered inapplicable. In several Asian countries, such as Japan and South Korea, the peak age of breast cancer incidence in women mainly ranges from 45 to 69 years old which is more than 10 years earlier than that in Europe and the United States [ 43 , 44 ]. Although some Asian guidelines agreed on beginning screening from the age of 40 or 45 years [ 9 , 30 , 32 ], high-quality evidence from large population-based RCTs is insufficient. In addition, based on several RCTs conducted in Canada, the UK, and Sweden, ECIBC and CTFPHC did not recommend regular screening begin at 40–44 years since the lower absolute benefit and higher overdiagnosis and false positives rate with related biopsies of this age group [ 2 , 25 , 27 , [45] , [46] , [47] ]. Furthermore, nine of 23 guidelines did not recommend an upper age limit. However, some guidelines recommended against regular screening for women older than age 70 or 75 years, as the harm potentially exceeds the benefits if screening is continued after these age groups [ 48 ]. The risk of breast cancer increases with age. Consequently, the decision to stop screening should be individually based on life expectancy or comorbid conditions.

Currently, MAM is widely accepted in developed countries with sufficient evidence to decrease breast cancer mortality among women aged 50–74 years and is recommended as a primary screening method in most screening guidelines [ 49 ]. Due to relatively high cost and the demand for high-quality radiologists, the application of MAM in low resource areas is limited [ 50 ]. Additionally, because higher mammographic density is associated with the masking of breast cancer on a mammogram, the sensitivity of MAM for women with dense breasts is lower than that for women with mainly fatty breasts [ 51 ]. Mammographic density among Asian women is higher than among Western women [ 52 ]. Several Asian studies have shown that US can improve the detection rate of breast cancer for women with dense breasts [ 53 , 54 ]. However, there is limited evidence for US in breast cancer screening to reduce mortality. Accordingly, the guidelines from European and American countries did not recommend US as the primary technique for breast cancer screening in average-risk population, but mainly as a supplemental method to MAM. Among the included guidelines of the present study, only Chinese guidelines recommended US as the primary screening tool. China has carried out a national breast cancer screening program since 2009. The screening tool of the program was changed from CBE to US in 2012, which provided preliminary evidence for the application of US in breast cancer screening in other Asian countries [ 55 , 56 ].

With greater emphasis on more accurate risk management based on patients and more personalized recommendations for diagnosis, treatment, and follow-up, age-oriented screening suggestions have been shifted to risk-based screening recommendations. By accurately identifying women who are above-average risk in the general population, we can provide timely and effective early diagnosis measures. High-risk women identified in the guidelines fell within many categories. The related recommendations for every category of high-risk women were different, which brought some difficulties to the implementation of breast cancer screening for high-risk women in the low resource areas. Thereby, identifying the risk factors of breast cancer by establishing a risk assessment model may be an effective way to prevent breast cancer. Currently, various risk prediction models were developed, such as the Gail model and BOADICEA model, whose application values in different countries are still under evaluation [ 57 , 58 ]. It is reported that China applies risk models as supplementary tools for screening in urban areas [ 59 ].

Few guidelines provided explicit recommendations for the management of women with positive findings except for NCCN [ 18 ] and NCC China [ 32 ]. Improper management of abnormal screening results may compromise the effectiveness of breast cancer screening programs. Doubeni et al. performed the PROSPR multi-model microsimulation study, which showed that the relative risk for the late-stage disease was higher when the time for diagnostic testing was delayed after an abnormal mammogram [ 60 ]. A previous study observed that low-income women and women of ethnic minority (African-American and Asian women) were less likely to have adequate follow-up abnormal breast cancer screening mammograms [ 61 ]. For these reasons, it is necessary to explore different referral and recall standards according to different initial screening results, to make a balance between the anxiety caused by false-positive breast cancer and the benefit of follow-up.

The strengths of this systematic review include its originality and the most comprehensive search strategy. This study was the largest and comprehensive systematic review to map the recommendations of the latest international breast screening guidelines. Furthermore, we systemically summarized the screening recommendations for both average-risk women and high-risk women.

Our study has some limitations. Even though we performed a comprehensive systematic search, we could not find all relevant guidelines. And we also did not include the breast screening program protocols in some countries. Another limitation was that non-English guidelines were not included in this review due to translation restrictions.

5. Conclusions

In summary, this study reviewed and compared the latest international breast screening guidelines for women both at average risk and at higher risk. The majority of guidelines were issued by developed countries, containing roughly the same but not identical recommendations for breast cancer on screening age, methods, and intervals. Most guidelines recommended annual or biennial mammographic screening for average-risk populations aged between 40 and 74 years and early annual MAM or annual MRI for high-risk populations. Current guidelines varied in methodological quality and increased efforts are needed to develop high-quality guidelines to provide more powerful supporting evidence for guidelines users. LMICs lacked published tailored clinical practice guideline. Therefore, we encourage policymakers and clinicians to use the evidence-based guidelines with higher AGREE Ⅱ scores considering local adaption.

Funding source

This work was supported by International Agency for Research on Cancer, France; World Health Organization, Switzerland [grant numbers CRA/SCR/2019/1].

Declaration of competing interest

All authors declare that they have no conflict of interest.

Acknowledgments

We gratefully acknowledge Ms. Huijiao Yan for her linguistic assistance during the revision of this manuscript.

☆ Present address: Department of Cancer Epidemiology, National Cancer Centre/National Clinical Research Centre for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 South Pan Jia Yuan Lane, Beijing 100,021, China.

Appendix A. Electronic search strategies

A) medline (via pubmed).

((Breast Neoplasms [MH] OR breast cancer* [tiab] OR breast neoplasm* [tiab] OR breast carcinoma* [tiab] OR breast tumor* [tiab] OR breast tumour* [tiab] OR mammary cancer* [tiab] OR mammary neoplasm* [tiab] OR mammary carcinoma* [tiab] OR mammary tumor* [tiab] OR mammary tumour* [tiab])

("Mass Screening" [Mesh] OR "Early Detection of Cancer" [Mesh] OR screening [tiab] OR early detect*[tiab])

(("Guideline" [Publication Type] OR "Practice Guideline" [Publication Type]) OR ("Guidelines as Topic" [Mesh] OR "Health Planning Guidelines" [Mesh] OR consensus [MeSH]) OR (guideline [Title] OR guidelines [Title] OR "practice guideline" [Title] OR "practice guidelines" [Title] OR "Health Planning Guidelines" [Title] OR Guidance [Title] OR consensus [Title] OR recommendations [Title] OR recommendation [Title] OR manual [Title] OR guidebook [Title] OR guidebooks [Title] OR guide [Title] OR guides [Title] OR handbook [Title] OR handbooks [Title])))

B) EMBASE via embase.com

('breast cancer'/exp OR 'breast tumor'/exp OR 'breast carcinoma*'/exp OR ('breast neoplasm*' OR 'breast tumor*' OR 'breast tumour*' OR 'mammary cancer*' OR 'mammary neoplasm*' OR 'mammary carcinoma*' OR 'mammary tumor*' OR 'mammary tumour*'):ab,ti

('Mass Screening'/exp OR 'early cancer diagnosis'/exp OR "screening":ab, ti OR early detect:ab,ti)

('Practice Guideline'/exp OR 'health care planning'/exp OR consensus/exp OR (guideline OR guidelines OR 'practice guideline' OR 'practice guidelines' OR 'Health Planning Guidelines' OR Guidance OR consensus OR recommendations OR recommendation OR manual OR guidebook OR guidebooks OR guide OR guides OR handbook OR handbooks):ti)

C) Web of Science

TI or AB=("breast cancer*" OR "breast neoplasm*" OR "breast carcinoma*" OR "breast tumor*" OR "breast tumour*" OR "mammary cancer*" OR "mammary neoplasm*" OR "mammary carcinoma*" OR "mammary tumor*" OR "mammary tumour*")

TI or AB=("Mass Screening" OR "Early Detection of Cancer" OR screening OR "early detect*")

TI=(guideline OR guidelines OR "Practice Guideline" OR "practice guidelines" OR consensus OR Guidance OR recommendation OR recommendations OR manual OR guide OR guides OR guidebook OR guidebooks OR handbook OR handbooks)

TITLE-ABS("breast cancer*" OR "breast neoplasm*" OR "breast carcinoma*" OR "breast tumor*" OR "breast tumour*" OR "mammary cancer*" OR "mammary neoplasm*" OR "mammary carcinoma*" OR "mammary tumor*" OR "mammary tumour*")

TITLE-ABS("Mass Screening" OR "Early Detection of Cancer" OR screening OR "early detect*")

TITLE (guideline OR "Practice Guideline" OR consensus OR Guidance OR recommendation OR manual OR guide OR guidebook OR handbook)

  • Conclusions
  • Article Information

Evidence reviews for the USPSTF use an analytic framework to visually display the key questions that the review will address in order to allow the USPSTF to evaluate the effectiveness and safety of a preventative service. The questions are depicted by linkages that relate interventions and outcomes. A dashed line indicates a health outcome that immediately follows an intermediate outcome. For additional details see the US Preventive Services Task Force Procedure Manual. 13

Reasons for exclusion: Design: Study did not use an included design. Outcomes: Study did not have relevant outcomes or had incomplete outcomes. Comparator: Study used an excluded comparator. Intervention: Study used an excluded intervention/screening approach. Population: Study was not conducted in an average-risk population. Timing: Study only reported first (prevalence) round screening follow-up. Publication type: Study was published in non–English-language or only available in an abstract. Quality: Study did not meet criteria for fair or good quality. Setting: Study was not conducted in a setting relevant to US practice. KQ indicates key question.

DBT indicates digital breast tomosynthesis; DM, digital mammography; and RR, relative risk.

a From random-effects restricted maximum likelihood model.

eMethods. Literature Search Strategies for Primary Literature

eTable 1. Inclusion and Exclusion Criteria

eTable 2. Quality Assessment Criteria

eTable 3. Included Studies and Their Ancillary Publications

eTable 4. Screen-Detected DCIS Diagnosed in Studies Comparing Digital Breast Tomosynthesis and Digital Mammography

eFigure 1. Pooled Analysis of Screen-Detected Invasive Cancers Diagnosed in Trials Comparing Digital Breast Tomosynthesis and Digital Mammography

eFigure 2. Pooled Analysis of Interval Cancers Diagnosed in Trials Comparing Digital Breast Tomosynthesis and Digital Mammography

eFigure 3. Cumulative Probability of False-Positive Biopsy in One NSRI Using BCSC Data Comparing Annual vs Biennial Screening with DBT or DM

eFigure 4. Cumulative Probability of False-Positive Recall in One NSRI Using BCSC Data Comparing Annual vs Biennial Screening with DBT or DM

eFigure 5. Cumulative Probability of False-Positive Recall or Biopsy in One NSRI Using BCSC Data Comparing Annual vs Biennial Screening with DBT or DM, among Women with Extremely Dense Breasts

  • USPSTF Recommendation: Screening for Breast Cancer JAMA US Preventive Services Task Force June 11, 2024 This 2024 Recommendation Statement from the US Preventive Services Task Force recommends biennial screening mammography for women aged 40 to 74 years (B recommendation) and concludes that evidence is insufficient to assess the balance of benefits and harms of screening mammography in women 75 years or older (I statement) and of screening using ultrasonography or MRI in women with dense breasts on a negative mammogram (I statement). US Preventive Services Task Force; Wanda K. Nicholson, MD, MPH, MBA; Michael Silverstein, MD, MPH; John B. Wong, MD; Michael J. Barry, MD; David Chelmow, MD; Tumaini Rucker Coker, MD, MBA; Esa M. Davis, MD, MPH; Carlos Roberto Jaén, MD, PhD, MS; Marie Krousel-Wood, MD, MSPH; Sei Lee, MD, MAS; Li Li, MD, PhD, MPH; Carol M. Mangione, MD, MSPH; Goutham Rao, MD; John M. Ruiz, PhD; James J. Stevermer, MD, MSPH; Joel Tsevat, MD, MPH; Sandra Millon Underwood, PhD, RN; Sarah Wiehe, MD, MPH
  • USPSTF Report: Collaborative Modeling to Compare Breast Cancer Screening Strategies JAMA US Preventive Services Task Force June 11, 2024 This modeling study uses Cancer Intervention and Surveillance Modeling Network models and national data on breast cancer incidence, mammography performance, treatment effects, and other-cause mortality in US women without previous cancer diagnoses to estimate outcomes of various mammography screening strategies. Amy Trentham-Dietz, PhD, MS; Christina Hunter Chapman, MD, MS; Jinani Jayasekera, PhD, MS; Kathryn P. Lowry, MD; Brandy M. Heckman-Stoddard, PhD, MPH; John M. Hampton, MS; Jennifer L. Caswell-Jin, MD; Ronald E. Gangnon, PhD; Ying Lu, PhD, MS; Hui Huang, MS; Sarah Stein, PhD; Liyang Sun, MS; Eugenio J. Gil Quessep, MS; Yuanliang Yang, MS; Yifan Lu, BASc; Juhee Song, PhD; Diego F. Muñoz, PhD; Yisheng Li, PhD, MS; Allison W. Kurian, MD, MSc; Karla Kerlikowske, MD; Ellen S. O’Meara, PhD; Brian L. Sprague, PhD; Anna N. A. Tosteson, ScD; Eric J. Feuer, PhD; Donald Berry, PhD; Sylvia K. Plevritis, PhD; Xuelin Huang, PhD; Harry J. de Koning, MD, PhD; Nicolien T. van Ravesteyn, PhD; Sandra J. Lee, ScD; Oguzhan Alagoz, PhD, MS; Clyde B. Schechter, MD, MA; Natasha K. Stout, PhD; Diana L. Miglioretti, PhD, ScM; Jeanne S. Mandelblatt, MD, MPH
  • Toward More Equitable Breast Cancer Outcomes JAMA Editorial June 11, 2024 Joann G. Elmore, MD, MPH; Christoph I. Lee, MD, MS
  • When Is It Best to Begin Mammograms, and How Often? JAMA Medical News & Perspectives June 11, 2024 This Medical News story discusses new USPSTF recommendations about the timing of screening mammograms. Rita Rubin, MA
  • Screening for Breast Cancer JAMA JAMA Patient Page June 11, 2024 In this JAMA Patient Page, the US Preventive Services Task Force provides a guide to screening for breast cancer. US Preventive Services Task Force
  • New Recommendations for Breast Cancer Screening—In Pursuit of Health Equity JAMA Network Open Editorial April 30, 2024 Lydia E. Pace, MD, MPH; Nancy L. Keating, MD, MPH
  • USPSTF Breast Cancer Screening Guidelines Do Not Go Far Enough JAMA Oncology Editorial June 1, 2024 Wendie A. Berg, MD, PhD

See More About

Select your interests.

Customize your JAMA Network experience by selecting one or more topics from the list below.

  • Academic Medicine
  • Acid Base, Electrolytes, Fluids
  • Allergy and Clinical Immunology
  • American Indian or Alaska Natives
  • Anesthesiology
  • Anticoagulation
  • Art and Images in Psychiatry
  • Artificial Intelligence
  • Assisted Reproduction
  • Bleeding and Transfusion
  • Caring for the Critically Ill Patient
  • Challenges in Clinical Electrocardiography
  • Climate and Health
  • Climate Change
  • Clinical Challenge
  • Clinical Decision Support
  • Clinical Implications of Basic Neuroscience
  • Clinical Pharmacy and Pharmacology
  • Complementary and Alternative Medicine
  • Consensus Statements
  • Coronavirus (COVID-19)
  • Critical Care Medicine
  • Cultural Competency
  • Dental Medicine
  • Dermatology
  • Diabetes and Endocrinology
  • Diagnostic Test Interpretation
  • Drug Development
  • Electronic Health Records
  • Emergency Medicine
  • End of Life, Hospice, Palliative Care
  • Environmental Health
  • Equity, Diversity, and Inclusion
  • Facial Plastic Surgery
  • Gastroenterology and Hepatology
  • Genetics and Genomics
  • Genomics and Precision Health
  • Global Health
  • Guide to Statistics and Methods
  • Hair Disorders
  • Health Care Delivery Models
  • Health Care Economics, Insurance, Payment
  • Health Care Quality
  • Health Care Reform
  • Health Care Safety
  • Health Care Workforce
  • Health Disparities
  • Health Inequities
  • Health Policy
  • Health Systems Science
  • History of Medicine
  • Hypertension
  • Images in Neurology
  • Implementation Science
  • Infectious Diseases
  • Innovations in Health Care Delivery
  • JAMA Infographic
  • Law and Medicine
  • Leading Change
  • Less is More
  • LGBTQIA Medicine
  • Lifestyle Behaviors
  • Medical Coding
  • Medical Devices and Equipment
  • Medical Education
  • Medical Education and Training
  • Medical Journals and Publishing
  • Mobile Health and Telemedicine
  • Narrative Medicine
  • Neuroscience and Psychiatry
  • Notable Notes
  • Nutrition, Obesity, Exercise
  • Obstetrics and Gynecology
  • Occupational Health
  • Ophthalmology
  • Orthopedics
  • Otolaryngology
  • Pain Medicine
  • Palliative Care
  • Pathology and Laboratory Medicine
  • Patient Care
  • Patient Information
  • Performance Improvement
  • Performance Measures
  • Perioperative Care and Consultation
  • Pharmacoeconomics
  • Pharmacoepidemiology
  • Pharmacogenetics
  • Pharmacy and Clinical Pharmacology
  • Physical Medicine and Rehabilitation
  • Physical Therapy
  • Physician Leadership
  • Population Health
  • Primary Care
  • Professional Well-being
  • Professionalism
  • Psychiatry and Behavioral Health
  • Public Health
  • Pulmonary Medicine
  • Regulatory Agencies
  • Reproductive Health
  • Research, Methods, Statistics
  • Resuscitation
  • Rheumatology
  • Risk Management
  • Scientific Discovery and the Future of Medicine
  • Shared Decision Making and Communication
  • Sleep Medicine
  • Sports Medicine
  • Stem Cell Transplantation
  • Substance Use and Addiction Medicine
  • Surgical Innovation
  • Surgical Pearls
  • Teachable Moment
  • Technology and Finance
  • The Art of JAMA
  • The Arts and Medicine
  • The Rational Clinical Examination
  • Tobacco and e-Cigarettes
  • Translational Medicine
  • Trauma and Injury
  • Treatment Adherence
  • Ultrasonography
  • Users' Guide to the Medical Literature
  • Vaccination
  • Venous Thromboembolism
  • Veterans Health
  • Women's Health
  • Workflow and Process
  • Wound Care, Infection, Healing

Others Also Liked

  • Download PDF
  • X Facebook More LinkedIn
  • CME & MOC

Henderson JT , Webber EM , Weyrich MS , Miller M , Melnikow J. Screening for Breast Cancer : Evidence Report and Systematic Review for the US Preventive Services Task Force . JAMA. 2024;331(22):1931–1946. doi:10.1001/jama.2023.25844

Manage citations:

© 2024

  • Permissions

Screening for Breast Cancer : Evidence Report and Systematic Review for the US Preventive Services Task Force

  • 1 Kaiser Permanente Evidence-based Practice Center, Center for Health Research, Portland, Oregon
  • 2 University of California Davis Center for Healthcare Policy and Research, Sacramento
  • Editorial Toward More Equitable Breast Cancer Outcomes Joann G. Elmore, MD, MPH; Christoph I. Lee, MD, MS JAMA
  • Editorial New Recommendations for Breast Cancer Screening—In Pursuit of Health Equity Lydia E. Pace, MD, MPH; Nancy L. Keating, MD, MPH JAMA Network Open
  • Editorial USPSTF Breast Cancer Screening Guidelines Do Not Go Far Enough Wendie A. Berg, MD, PhD JAMA Oncology
  • US Preventive Services Task Force USPSTF Recommendation: Screening for Breast Cancer US Preventive Services Task Force; Wanda K. Nicholson, MD, MPH, MBA; Michael Silverstein, MD, MPH; John B. Wong, MD; Michael J. Barry, MD; David Chelmow, MD; Tumaini Rucker Coker, MD, MBA; Esa M. Davis, MD, MPH; Carlos Roberto Jaén, MD, PhD, MS; Marie Krousel-Wood, MD, MSPH; Sei Lee, MD, MAS; Li Li, MD, PhD, MPH; Carol M. Mangione, MD, MSPH; Goutham Rao, MD; John M. Ruiz, PhD; James J. Stevermer, MD, MSPH; Joel Tsevat, MD, MPH; Sandra Millon Underwood, PhD, RN; Sarah Wiehe, MD, MPH JAMA
  • US Preventive Services Task Force USPSTF Report: Collaborative Modeling to Compare Breast Cancer Screening Strategies Amy Trentham-Dietz, PhD, MS; Christina Hunter Chapman, MD, MS; Jinani Jayasekera, PhD, MS; Kathryn P. Lowry, MD; Brandy M. Heckman-Stoddard, PhD, MPH; John M. Hampton, MS; Jennifer L. Caswell-Jin, MD; Ronald E. Gangnon, PhD; Ying Lu, PhD, MS; Hui Huang, MS; Sarah Stein, PhD; Liyang Sun, MS; Eugenio J. Gil Quessep, MS; Yuanliang Yang, MS; Yifan Lu, BASc; Juhee Song, PhD; Diego F. Muñoz, PhD; Yisheng Li, PhD, MS; Allison W. Kurian, MD, MSc; Karla Kerlikowske, MD; Ellen S. O’Meara, PhD; Brian L. Sprague, PhD; Anna N. A. Tosteson, ScD; Eric J. Feuer, PhD; Donald Berry, PhD; Sylvia K. Plevritis, PhD; Xuelin Huang, PhD; Harry J. de Koning, MD, PhD; Nicolien T. van Ravesteyn, PhD; Sandra J. Lee, ScD; Oguzhan Alagoz, PhD, MS; Clyde B. Schechter, MD, MA; Natasha K. Stout, PhD; Diana L. Miglioretti, PhD, ScM; Jeanne S. Mandelblatt, MD, MPH JAMA
  • Medical News & Perspectives When Is It Best to Begin Mammograms, and How Often? Rita Rubin, MA JAMA
  • JAMA Patient Page Screening for Breast Cancer US Preventive Services Task Force JAMA

Importance   Breast cancer is a leading cause of cancer mortality for US women. Trials have established that screening mammography can reduce mortality risk, but optimal screening ages, intervals, and modalities for population screening guidelines remain unclear.

Objective   To review studies comparing different breast cancer screening strategies for the US Preventive Services Task Force.

Data Sources   MEDLINE, Cochrane Library through August 22, 2022; literature surveillance through March 2024.

Study Selection   English-language publications; randomized clinical trials and nonrandomized studies comparing screening strategies; expanded criteria for screening harms.

Data Extraction and Synthesis   Two reviewers independently assessed study eligibility and quality; data extracted from fair- and good-quality studies.

Main Outcomes and Measures   Mortality, morbidity, progression to advanced cancer, interval cancers, screening harms.

Results   Seven randomized clinical trials and 13 nonrandomized studies were included; 2 nonrandomized studies reported mortality outcomes. A nonrandomized trial emulation study estimated no mortality difference for screening beyond age 74 years (adjusted hazard ratio, 1.00 [95% CI, 0.83 to 1.19]). Advanced cancer detection did not differ following annual or biennial screening intervals in a nonrandomized study. Three trials compared digital breast tomosynthesis (DBT) mammography screening with digital mammography alone. With DBT, more invasive cancers were detected at the first screening round than with digital mammography, but there were no statistically significant differences in interval cancers (pooled relative risk, 0.87 [95% CI, 0.64-1.17]; 3 studies [n = 130 196]; I 2  = 0%). Risk of advanced cancer (stage II or higher) at the subsequent screening round was not statistically significant for DBT vs digital mammography in the individual trials. Limited evidence from trials and nonrandomized studies suggested lower recall rates with DBT. An RCT randomizing individuals with dense breasts to invitations for supplemental screening with magnetic resonance imaging reported reduced interval cancer risk (relative risk, 0.47 [95% CI, 0.29-0.77]) and additional false-positive recalls and biopsy results with the intervention; no longer-term advanced breast cancer incidence or morbidity and mortality outcomes were available. One RCT and 1 nonrandomized study of supplemental ultrasound screening reported additional false-positives and no differences in interval cancers.

Conclusions and Relevance   Evidence comparing the effectiveness of different breast cancer screening strategies is inconclusive because key studies have not yet been completed and few studies have reported the stage shift or mortality outcomes necessary to assess relative benefits.

Breast cancer is the second leading cause of cancer mortality for US women, despite a steady overall decline in breast-cancer mortality rates over the past 20 years. 1 The average age-adjusted rate for the years 2016-2020 was 19.6 per 100 000, with an estimated 43 170 deaths in 2023. 1 , 2 The majority of cases occur between the ages of 55 and 74 years, 1 and incidence is highest among women ages 70 to 74 (468.2 per 100 000). 3 Non-Hispanic White women have the highest breast cancer incidence, 4 but mortality is 40% higher for non-Hispanic Black women (27.6 per 100 000) compared with White women (19.7 per 100 000); non-Hispanic Black women experience lower 5-year survival regardless of the cancer subtype or stage at the time of detection. 1 , 5 - 7

Previous reviews of breast cancer screening effectiveness established the benefits and harms of mammography based primarily on large, long-term trials. 8 , 9 In 2016, the US Preventive Services Task Force (USPSTF) recommended screening for breast cancer in women starting at age 50 years every 2 years continuing through age 74 years (B recommendation) and that screening from ages 40 to 49 years should be based on clinical discussions of patient preferences and individual breast cancer risk (C recommendation). 10 This comparative effectiveness systematic review of breast cancer screening strategies was conducted concurrently with a separate decision modeling study. 11 Both informed the USPSTF updated breast cancer screening recommendations. 12

This review addressed 3 key questions (KQs) on the comparative effectiveness and harms of different screening strategies ( Figure 1 ). Methodological details including study selection, a list of excluded studies, detailed study-level results for all outcomes and for specific subpopulations, and contextual observations are available in the full evidence report. 14

Studies included in the 2016 USPSTF reviews 8 , 9 , 15 , 16 were evaluated for inclusion with eligibility criteria for the current review. In addition, database searches for relevant studies published between January 2014 and August 22, 2022, were conducted in MEDLINE, the Cochrane Central Register of Controlled Clinical Trials, and the Cochrane Database of Systematic Reviews (eMethods in the Supplement ). Reference lists of other systematic reviews were searched to identify additional relevant studies. ClinicalTrials.gov was searched for relevant ongoing trials. Ongoing surveillance to identify newly published studies was conducted through March 2024 to identify major studies published in the interim. Two new nonrandomized studies were identified 17 , 18 and are not further discussed, as they would not change interpretation of the review findings or conclusions.

Two independent reviewers screened titles, abstracts, and relevant full-text articles to ensure consistency with a priori inclusion and exclusion criteria (eTable 1 in the Supplement ). We included English-language studies of asymptomatic screening populations not at high risk for breast cancer. The eligible population for this review is adult females (sex assigned at birth). For consistency with the underlying evidence, the term “women” is used throughout this report; however, cancer registries and studies of breast cancer generally infer gender based on physiology and medical history rather than measuring self-reported gender. Included studies compared mammography screening modalities (mammography with or without digital breast tomosynthesis [DBT]), different screening strategies with respect to interval, age to start, age to stop, or supplemental screening strategies using ultrasound or magnetic resonance imaging (MRI) with mammography.

For KQ1, randomized clinical trials (RCTs) or nonrandomized studies of interventions with contemporaneous comparison groups that reported breast cancer morbidity, mortality, all-cause mortality, or quality of life were included. For KQ2, the primary outcome of interest was progression to advanced breast cancer, defined for this review as stage IIB or higher, which encompasses tumors with local lymph node involvement or distant metastases. 19 Study-defined advanced breast cancer outcomes were used when this outcome was not reported (eg, stage II or higher). Invasive breast cancer detection outcomes from multiple screening rounds can indicate whether a screening modality or strategy reduces the risk of advanced cancer by detecting early cancers that would otherwise have progressed (stage shift), thereby potentially reducing breast cancer morbidity and mortality. 20 - 23

For KQ3, RCTs and nonrandomized studies of interventions reporting adverse events, including psychological harms, radiation exposure, and interval invasive cancers (incident or missed due to false-negative screening) were included, regardless of the number of screening rounds reported. False-positive recall, false-positive biopsy recommendation, and false-positive biopsy rates (individuals who underwent a biopsy for a benign lesion) were obtained from included RCTs and from nonrandomized studies reporting cumulative rates of these potential harms of screening.

Two reviewers evaluated all articles that met inclusion criteria using prespecified quality criteria (eTable 2 in the Supplement ). Discordant quality ratings were resolved through discussion and input from a third reviewer. Risk-of-bias assessment was conducted using the USPSTF-specific criteria for randomized trials 13 and an adapted tool from the Risk of Bias in Non-Randomized Studies of Interventions (ROBINS-I). 24 Studies determined to be at high risk of bias were excluded. One reviewer extracted key elements of included studies into standardized evidence tables in DistillerSR (Evidence Partners) and a second reviewer checked the data for accuracy. Limited evidence on sub-KQs is available in the full report. 14 When available, reported relative risks were provided in the tables, but we calculated and reported crude effect estimates and confidence intervals when studies did not provide them. For KQ2 intermediate detection outcomes, the definition of advanced cancer reported in the studies was used for synthesis; commonly this was stage II or later. Comparisons of prognostic characteristics or markers (eg, grade, tumor size, nodal involvement, receptor status) were included for comparisons as data allowed.

All quantitative analyses were conducted in Stata version 16 (StataCorp). The presence of statistical heterogeneity was assessed among pooled studies using the I 2 statistic. Where effects were sufficiently consistent and clinical and statistical heterogeneity low, random-effects meta-analyses were conducted using the restricted maximum likelihood; all tests were 2-sided, with P  < .05 indicating statistical significance.

Aggregate strength of evidence (ie, high, moderate, or low) was assessed for each KQ and comparison using the approach described in the Methods Guide for the Effectiveness and Comparative Effectiveness Reviews, 25 based on consistency, precision, publication bias, and study quality.

Investigators reviewed 10 378 unique citations and 419 full-text articles for all KQs ( Figure 2 ). Twenty studies reported in 45 publications were included. 26 - 45 A full list of included studies by KQ is located in eTable 3 in the Supplement .

Key Question 1. What is the comparative effectiveness of different mammography-based breast cancer screening strategies (eg, by modality, interval, initiation and stopping age, use of supplemental imaging, or personalization based on risk factors) on breast cancer morbidity and mortality?

Two nonrandomized studies reported on the association of different screening programs with breast cancer morbidity and mortality. One study was designed to compare different ages to stop screening 30 and another compared annual and triennial screening intervals. 41

A fair-quality observational study (n = 1 058 013) on age to stop screening used an emulated trial methodology to analyze a random sample of US Medicare A and B claims data for enrollees aged 70 to 84 years (1999 to 2008), eligible for breast cancer screening, and with at least a 10-year estimated life expectancy. The study estimated the effect of stopping screening at ages 70, 75, and 80 years compared with continued annual screening. 30 , 46 Continuation of screening between the ages of 70 and 74 years was associated with reduced mortality risk based on survival analysis (hazard ratio, 0.78 [95% CI, 0.63 to 0.95]), but the absolute difference in the risk of death for the age group was small and the confidence interval included null (1.0 fewer deaths per 1000 screened [95% CI, −2.3 to 0.1]). These results indicate a difference in the cumulative incidence curves that approached a difference in the mortality risk for the age group. Conversely, continued screening vs no screening from ages 75 to 84 years did not result in statistically significant differences in the absolute risk of breast cancer mortality (0.07 fewer deaths per 1000 [95% CI, –0.93 to 1.3]) or the cumulative mortality incidence (hazard ratio, 1.00 [95% CI, 0.83 to 1.19]).

A fair-quality nonrandomized clinical study (n = 14 765) conducted in Finland during the years 1985 to 1995 assigned participants aged 40 to 49 years to annual or triennial screening invitations by alternating birth year. 41 The study reported no difference in breast cancer mortality: 20.3 deaths per 100 000 person-years with annual screening invitations and 17.9 deaths per 100 000 person-years with triennial screening invitations (relative risk [RR], 1.14 [95% CI, 0.59-1.27]).

Key Question 2. What is the comparative effectiveness of different mammography-based breast cancer screening strategies (eg, by modality, interval, initiation and stopping age, use of supplemental imaging, or personalization based on risk factors) on the incidence of and progression to advanced breast cancer?

No eligible studies of age to start or stop screening, supplemental screening, or personalized screening were included, because no RCTs or nonrandomized studies reported more than a single round of screening comparing screening strategies. For screening interval, 1 RCT 26 and 1 nonrandomized study, 41 and for comparisons of different screening modalities (DBT vs digital mammography) 3 RCTs 27 , 33 , 42 and 2 nonrandomized studies, 34 , 44 met eligibility criteria.

Two fair-quality studies addressed the effect of screening interval on the characteristics of detected cancers. A fair-quality United Kingdom Co-ordinating Committee on Cancer Research (UKCCCR) RCT comparing screening intervals was conducted as part of the UK National Breast Screening Program. The study randomized participants aged 50 to 62 years to annual (n = 37 530) or triennial (n = 38 492) breast cancer screening during the years 1989 to 1996. 26 After 3 years of screening (1 incidence screen in the triennial screening group), a similar number of cancers (screen-detected and interval) had been diagnosed in the annual and triennial screening groups (6.26 and 5.40 per 1000 screened, respectively; RR, 1.16 [95% CI, 0.96 to 1.40]). No statistically significant differences were found in the cancer characteristics (tumor size, nodal status, histological grade) between groups over the course of the study.

A fair-quality nonrandomized study using Breast Cancer Surveillance Consortium (BCSC) registry data (1996 to 2012) 39 found the relative risk of being diagnosed with a breast cancer with less favorable prognostic characteristics (stage IIB or higher, tumor size >15 mm, or node-positive) was not statistically different for women screened biennially compared with those screened annually for any age category (40-49, 50-59, 60-69, 70-85 years).

Three fair-quality RCTs 27 , 33 , 42 reported cancer detection over 2 rounds of screening, comparing the effects of screening with DBT and digital mammography on the presence of advanced cancer at subsequent screening rounds ( Table 1 ). Participants were randomized to the DBT intervention group or the digital mammography control group at a first round of screening, followed in 2 trials by a second round of screening with digital mammography for all second-round participants (Proteus Donna, 27 RETomo 42 ) and in 1 trial with DBT for all second-round participants (To-Be 33 ). The trials used an identical screening modality for both study groups at the second round because using the same instrument is a stronger design for detection of stage shift.

The RCTs reported increased detection of invasive cancer with DBT at the first round of screening (pooled RR, 1.41 [95% CI, 1.20 to 1.64]; 3 RCTs [n = 129 492]; I 2  = 7.6%) and no statistical difference in invasive cancer at the subsequent screening (pooled RR, 0.87 [95% CI, 0.73 to 1.05]; 3 RCTs [n = 105 064]; I 2  = 0%) (eFigure 1 in the Supplement ). 27 , 33 , 42 There was no statistically significant difference in the incidence of advanced cancers at the subsequent screening round (progression of cancers not found at prior screening that would indicate stage shift) in the individual trials ( Figure 3 ). Results were inconsistent and thus not pooled for the advanced cancer, larger tumor (>20 mm), and node-positive cancer outcomes. The results for histologic grade 3 cancer at the second screening were consistent (pooled RR, 0.97 [95% CI, 0.61-1.55]; 3 RCTs [n = 105 244]; I 2  = 0%) ( Figure 3 ). Due to the small number of cases, it was not possible to assess differences in the detection of cancers lacking hormone or growth factor receptors (ie, triple-negative cancers) that have the worst prognosis among breast cancer subtypes.

Two fair-quality nonrandomized studies of interventions (NRSIs), including a US study using BCSC data, compared breast cancer detection outcomes from screening over multiple rounds (≥2) with either DBT-based mammography or digital mammography alone. 34 , 44 The findings were generally consistent with the trial results for cancer detection and stage shift.

Key Question 3. What are the comparative harms of different mammography-based breast cancer screening strategies (modality, interval, initiation age, use of supplemental imaging, or personalization based on risk factors)?

No eligible studies of age to start screening or personalized screening were identified. For age to stop screening, 1 fair-quality nonrandomized study met eligibility criteria. 30 For comparisons of potential harms associated with different screening intervals, a fair-quality RCT 26 and 2 fair-quality nonrandomized studies 39 , 41 were included. For comparisons of different screening modalities (DBT vs digital mammography), 4 RCTs (3 good- and 1 fair-quality) 27 , 31 , 33 , 42 and 7 fair-quality nonrandomized studies were included. 28 , 32 , 34 - 36 , 43 , 44

In the NRSI using an emulated trial methodology to evaluate the age to stop screening, 30 the 8-year cumulative proportion of participants with a breast cancer diagnosis was higher among those who continued annual screening from ages 70 to 84 years (5.5%) compared with those who discontinued screening (3.9%) at age 70 years. Because fewer cancers were diagnosed among those who discontinued screening, there was a lower risk of undergoing cancer treatment and experiencing related morbidity. Notably, for participants aged 75 to 84 years, screening (and treatment) were not associated with lower breast cancer mortality (see KQ1 results).

The UKCCCR trial included for KQ2 26 reported fewer interval cancers (false-negative and incident cancers) diagnosed in the annual invitation group compared with triennial screening (1.84 vs 2.70 per 1000 women screened, respectively; RR, 0.68 [95% CI, 0.50 to 0.92]). The nonrandomized clinical trial conducted in Finland included for KQ1 41 also reported interval cancers diagnosed with annual vs triennial screening and found no statistical difference in incidence ( P  = .22, data not reported). Data from 2 studies from the BCSC registry reported higher probabilities of false-positive recalls and biopsy recommendations with annual screening compared with biennial screening and no statistical difference in interval cancers in adjusted analyses. 32 , 39 , 44

Four RCTs (3 good-quality, 1 fair-quality) 27 , 31 , 33 , 42 and 7 fair-quality nonrandomized studies 28 , 32 , 34 - 36 , 43 , 44 reported outcomes related to potential screening harms associated with DBT-based screening compared with digital mammography–only screening, including interval cancer rates, round-specific and cumulative false-positive recalls and biopsies, and radiation exposure. Meta-analysis of 3 large trials did not show a statistically significant difference in rates of interval cancer after screening with DBT compared with digital mammography (pooled RR, 0.87 [95% CI, 0.64 to 1.17]; 3 RCTs [n = 130 196]; I 2  = 0%) (eFigure 2 in the Supplement ). 27 , 33 , 42

Data on interval cancers were also obtained from 7 nonrandomized studies. 28 , 32 , 34 - 36 , 43 , 44 The most recent BCSC analysis, reporting interval cancer rates across multiple screening rounds with either DBT or digital mammography, did not identify statistically significant differences in invasive or advanced interval cancers. 44

The effects of DBT screening on false-positive recall and false-positive biopsy rates varied across studies 27 , 33 , 42 and by screening round, with small or no statistical differences between study groups, not consistently favoring DBT-based mammography or digital mammography.

Evidence from 2 nonrandomized BCSC studies provided false-positive results across several screening rounds. 32 , 44 In 1 study, rates of false-positive recall and false-positive biopsy rates were lower with DBT in initial screening rounds, but differences were attenuated and not statistically significant compared with digital mammography only after additional rounds of screening ( Table 2 ). 44 The other study reported no statistical difference in 10-year cumulative false-positive biopsy recommendation rates between biennial DBT and digital mammography screening, but false-positive recall was slightly lower with DBT (eFigures 3 and 4 in the Supplement ); no differences by modality were identified for individuals with extremely dense breasts in stratified analyses (eFigure 5 in the Supplement ). 32

Four RCTs 27 , 31 , 33 , 42 and 1 NRSI 35 reported the mean, median, or relative radiation dose received in each study group at a single screening round. The 3 studies using DBT/digital mammography screening reported radiation exposure approximately 2 times higher in the intervention group compared with the digital mammography–only group. 27 , 35 , 42 Differences between study groups in radiation exposure were smaller in studies using DBT with synthetic digital mammography. 33 , 47

The Dense Tissue and Early Breast Neoplasm Screening (DENSE) trial, a good-quality RCT conducted in the Netherlands, randomized (1:4) participants aged 50 to 75 years with extremely dense breasts and negative mammography findings (2011-2015) (n = 40 373) to an invitation or no invitation for supplemental MRI screening. 45 (The RCT was not included for KQ2 because second round results in the control group were unavailable). Fifty-nine percent of those randomized to the invitation underwent an MRI examination (n = 4783). In intention-to-treat analysis, 2.2 per 1000 experienced interval breast cancer diagnoses in the supplemental screening invitation group, compared with 4.7 per 1000 screened in the digital mammography control group (RR, 0.47 [95% CI, 0.29 to 0.77]). Adverse events related to the supplemental MRI screening reported in the trial included 5 classified as serious adverse events (2 vasovagal reactions and 3 allergic reactions to the contrast agent) and 2 reports of extravasation (leaking) of the contrast agents and 1 shoulder subluxation. Twenty-seven participants (0.6% of the MRI group) reported a serious adverse event within 30 days of the MRI. Those who underwent supplemental MRI screening also experienced additional recalls (94.9 per 1000 screened), false-positive recalls (80.0 per 1000 screened), and false-positive biopsies (62.7 per 1000 screened).

A fair-quality nonrandomized study used claims data from commercially insured women (MarketScan database) aged 40 to 64 years who had received at least 1 bilateral screening breast MRI (n = 9208) or mammogram (n = 9208) between January 2017 and June 2018. 29 Following propensity score matching, those undergoing screening with MRI were more likely to have additional health care cascade events such as office visits and follow-up tests unrelated to breast conditions (adjusted difference between groups, 19.6 per 100 screened [95% CI, 8.6 to 30.7]) in the subsequent 6 months.

A fair-quality RCT, the Japan Strategic Anti-cancer Randomized Trial, randomly assigned asymptomatic women aged 40 to 49 years (2007-2011) to breast cancer screening with mammography plus handheld ultrasound (digital mammography/ultrasound) (n = 36 859) or mammography only (digital mammography) (n = 36 139). 40 The relative risk of invasive interval cancer was not statistically significantly different for digital mammography/ultrasound vs digital mammography only (RR, 0.58 [95% CI, 0.31 to 1.08]). This result differs from the statistically significant population-average effect reported in the study ( P  = .03), which included interval ductal carcinoma in situ (proportion difference, −0.05% [95% CI, −0.09 to 0]). Those undergoing ultrasound in addition to digital mammography experienced 48.0 per 1000 additional false-positive recall results compared with those assigned to digital mammography screening only.

A fair-quality nonrandomized study using data from 2 BCSC registry sites compared screening outcomes for participants receiving ultrasonography on the same day as a screening mammogram (digital mammography/ultrasound) (n = 3386, contributing 6081 screens) compared with those that received only a mammogram (digital mammography) (n = 15 176, contributing 30 062 screens). 37 However, 31% of participants had a first-degree family history of breast cancer or previous breast biopsy. There was no statistical difference in interval cancer risk (adjusted RR, 0.67 [95% CI, 0.33 to 1.37]), and rates of false-positive biopsy were twice as high for the mammography/ultrasound group (adjusted RR, 2.23 [95% CI, 1.03 to 2.58]).

Prior screening effectiveness reviews based on large trials initiated in previous decades established a statistically significant mortality benefit for mammography screening of women aged 50 to 69 years. 8 , 9 , 15 The current review considered comparative effectiveness questions on the relative benefits and harms of different screening start and stop ages, intervals, and modalities for women at average breast cancer risk. Findings are summarized in Table 3 .

The evidence was insufficient for addressing the age to start or end screening. No eligible studies comparing different ages to start screening were identified. Limited evidence from 1 nonrandomized study, using an emulated trial study design, suggested that screening beyond age 74 years may not reduce breast cancer mortality. 30

Evidence was also insufficient for evaluating the effect of screening intervals on breast cancer morbidity and mortality. Two nonrandomized studies found no difference in breast cancer outcomes. 26 , 39 Moderate evidence supported longer screening intervals (eg, biennial) to reduce the cumulative risk of false-positive recall and biopsy. The observational studies of different screening intervals compared individuals who self-selected or were referred for different screening intervals, contributing to risk of bias in the results.

Results from 3 RCTs 27 , 33 , 42 and 2 nonrandomized studies 34 , 44 provided moderate evidence that DBT-based mammography does not reduce the risk of invasive interval cancer or advanced cancer at subsequent screening rounds. Additional rounds of screening and longer follow-up are needed to fully evaluate whether DBT reduces breast cancer morbidity and mortality. Consistent with trial findings, a nonrandomized BCSC study did not find reduced risks of advanced or interval cancers with DBT. 44 Limited evidence from trials on harms of screening with DBT 27 , 33 , 42 indicated similar false-positive recall and biopsy rates. An observational BCSC study did not show differences in the 10-year cumulative false-positive biopsy rates 32 ; lower false-positive recall and biopsy with DBT screening were attenuated after several screening rounds. 44 Additional research is needed to ascertain whether DBT-based screening would reduce false-positives over a lifetime of screening.

The evidence was not adequate to evaluate the benefits and harms of supplemental MRI screening for people with dense breasts. No eligible studies were identified that provide evidence on breast cancer morbidity or mortality outcomes with supplemental MRI screening compared with mammography alone among individuals with dense breasts. The DENSE trial 45 reported fewer interval cancers with 1 round of supplemental MRI screening, but results from a second screening round are not yet published. Evidence of higher advanced cancer incidence in the mammography-only group relative to the MRI group would be needed to anticipate effects on morbidity or mortality. Supplemental MRI led to additional false-positive recalls and biopsies, and uncommon but serious adverse events were observed. 45 Two recent systematic reviews of the test performance literature reported higher cancer detection with supplemental MRI screening along with substantially increased recall and biopsy rates among individuals without cancer. 48 , 49

Lack of a standardized and reliable assessment tool for measuring breast density and density variation across the lifespan pose challenges for research into the optimal screening strategy for persons with dense breasts. 16 Research is also needed to evaluate personalized risk-based screening, based on breast cancer risk factors and personal screening preferences. The ongoing WISDOM trial and My Personalized Breast Screening study (expected completion in 2025) may help to address these research gaps. 50 , 51

Breast cancer is an active area of research, yet few longitudinal RCTs comparing different screening strategies have been conducted following completion of the major trials that established the effectiveness of mammography for reducing breast cancer mortality for women aged 50 to 69 years. This review included 6 new randomized trials, 27 , 31 , 33 , 40 , 42 , 45 4 comparing DBT with digital mammography screening 27 , 31 , 33 , 42 and 2 on supplemental screening compared with mammography alone. 40 , 45 Three of these trials are ongoing 31 , 40 , 45 and have reported preliminary results only. Observational studies were also included, but few studies were available that followed up a screening population over time to compare the health outcomes associated with different screening approaches. These studies, while potentially more representative of a screening population, have higher risk of biased results due to confounding and selection.

Changes in population health, imaging technologies, and available treatments may limit the applicability of previous studies. Recent trials included in this review were conducted outside of the US and enrolled mostly White European populations. No studies evaluated screening outcomes for racial or ethnic groups in the US that experience health inequities and higher rates of breast cancer mortality. Black women are at highest risk of breast cancer mortality, 52 with lower 5-year survival than all other race and ethnicity groups. 7 Breast cancer mortality risk also increases at younger ages for Black women compared with White women. 53 This review did not address additional factors beyond screening that contribute to breast cancer mortality inequities. 54 Rigorous research is essential to understand and identify improvements needed along the pathway from screening to treatment 55 and to address inequities in follow-up time after a positive screening result, time to diagnosis, 56 - 60 and receipt of high-quality treatment and support services. 59 , 61 , 62

Evidence comparing outcomes for different screening intervals and ages to start and stop screening was limited or absent. Trials of personalized screening based on risk and patient preferences are in progress and may address evidence gaps related to optimal screening start ages and intervals. Research is needed to better characterize potential harms of screening, including patient perspectives on experiencing false-positive screening results. Women with false-positive screening results may be less likely to return for their next scheduled mammogram, as reported in a large US health system study. 55 , 63 Rigorous studies that enroll screening populations and report advanced cancer detection, morbidity, and mortality outcomes from multiple rounds of screening are needed to overcome persistent limitations in the evidence on breast cancer screening. Multiple screening rounds are essential to determine whether a screening modality or strategy reduces the risk of advanced cancer by detecting early cancers that would otherwise have progressed (stage shift), potentially reducing breast cancer morbidity and mortality. 20 - 23 , 64

The potential benefits of risk-stratified screening strategies, including the use of supplemental screening with ultrasound or MRI, have not been fully evaluated, although some harms are evident. Longer term follow-up on existing comparative effectiveness trials, complete results from ongoing RCTs of personalized screening programs, 65 , 66 and rigorous new studies are needed to further strengthen the evidence and optimize breast cancer screening strategies.

Evidence comparing the effectiveness of different breast cancer screening strategies is inconclusive because key studies have not yet been completed and few studies have reported the stage shift or mortality outcomes necessary to assess relative benefits.

Accepted for Publication: November 23, 2023.

Published Online: April 30, 2024. doi:10.1001/jama.2023.25844

Corresponding Author: Jillian T. Henderson, PhD, MPH, Kaiser Permanente Evidence-based Practice Center, Center for Health Research, Kaiser Permanente Northwest, 3800 N Interstate Ave, Portland, OR 97227 ( [email protected] ).

Author Contributions: Dr Henderson had full access to all of the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

Concept and design: All authors.

Acquisition, analysis, or interpretation of data: All authors.

Drafting of the manuscript: All authors.

Critical review of the manuscript for important intellectual content: Henderson, Weyrich, Miller.

Statistical analysis: Henderson.

Administrative, technical, or material support: Webber, Melnikow.

Supervision: Henderson.

Conflict of Interest Disclosures: None reported.

Funding/Support: This research was funded under contract number 75Q80120D00004, Task Order 75Q80121F32004, from the Agency for Healthcare Research and Quality (AHRQ), US Department of Health and Human Services.

Role of the Funder/Sponsor: Investigators worked with USPSTF members and AHRQ staff to develop the scope, analytic framework, and key questions for this review. AHRQ had no role in study selection, quality assessment, or synthesis. AHRQ staff provided project oversight, reviewed the report to ensure that the analysis met methodological standards, and distributed the draft for peer review. Otherwise, AHRQ had no role in the conduct of the study; collection, management, analysis, and interpretation of the data; and preparation, review, or approval of the manuscript findings.

Disclaimer: The opinions expressed in this document are those of the authors and do not reflect the official position of AHRQ or the US Department of Health and Human Services.

Additional Contributions: The authors gratefully acknowledge the following individuals for their contributions to this project: Howard Tracer, MD (AHRQ); Heidi D. Nelson, MD, MPH, MACP (Kaiser Permanente Bernard J. Tyson School of Medicine); current and former members of the USPSTF who contributed to topic deliberations; and Evidence-based Practice Center staff members Melinda Davies, MA, Jill Pope, and Leslie A. Purdue, MPH, for technical and editorial assistance at the Kaiser Permanente Center for Health Research. USPSTF members, peer reviewers, and federal partner reviewers did not receive financial compensation for their contributions.

Additional Information: A draft version of this evidence report underwent external peer review from 5 content and methods experts (Nehmat Houssami, MBBS, MPH, Med, PhD [University of Sydney-Australia]; Patricia Ganz, MD [UCLA]; Gerald Gartlehner, MD, MPH [Cochrane Austria]; Karla Kerlikowske, MD [UC San Francisco]; Lisa Newman, MD, MPH [New York Presbyterian/Weill Cornell Medical Center]) and 4 scientific representatives from 3 federal partner organizations (Centers for Disease Control and Prevention; Office of Research on Women’s Health; National Institute on Minority Health and Health Disparities). Comments were presented to the USPSTF during its deliberation of the evidence and were considered in preparing the final evidence review.

Editorial Disclaimer: This evidence report is presented as a document in support of the accompanying USPSTF Recommendation Statement. It did not undergo additional peer review after submission to JAMA .

  • Register for email alerts with links to free full-text articles
  • Access PDFs of free articles
  • Manage your interests
  • Save searches and receive search alerts

COMMENTS

  1. A Systematic Literature Review of Breast Cancer Diagnosis ...

    In this paper, different machine intelligence techniques [machine learning (ML), and deep learning (DL)] were analysed in the context of breast cancer. In addition, the classification of breast cancer into malignant and benign using different breast cancer image modalities were discussed.

  2. A Systematic Literature Review of Health Utility Values in ...

    This systematic literature review identified the full range of published health utility values relevant to breast cancer from diagnostic or screening, local and systemic therapies, allied health or complementary medicine–related interventions, and treatment-related adverse events.

  3. Breast Cancer—Epidemiology, Risk Factors, Classification ...

    Abstract. Breast cancer (BC) is the most frequently diagnosed cancer in women worldwide with more than 2 million new cases in 2020. Its incidence and death rates have increased over the last three decades due to the change in risk factor profiles, better cancer registration, and cancer detection.

  4. Breast cancer diagnosis: A systematic review - ScienceDirect

    Mammography images, breast thermal images, and breast ultrasound images are the three ways to diagnose breast cancer. The paper will discuss some recent developments in machine learning and deep learning in three different breast cancer diagnosis methods.

  5. Global guidelines for breast cancer screening: A systematic ...

    Abstract. Objectives. Breast cancer screening guidelines could provide valuable tools for clinical decision making by reviewing the available evidence and providing recommendations. Little information is known about how many countries have issued breast cancer screening guidelines and the differences among existing guidelines.

  6. Screening for Breast Cancer Evidence Report and Systematic ...

    Invasive breast cancer detection outcomes from multiple screening rounds can indicate whether a screening modality or strategy reduces the risk of advanced cancer by detecting early cancers that would otherwise have progressed (stage shift), thereby potentially reducing breast cancer morbidity and mortality. 20 - 23.